Language selection

Search

Patent 2964764 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2964764
(54) English Title: COMBINATION THERAPY FOR USE IN CANCER THERAPY
(54) French Title: POLYTHERAPIE DESTINEE A ETRE UTILISEE EN CANCEROTHERAPIE
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 1/68 (2018.01)
(72) Inventors :
  • PATERSON, YVONNE (United States of America)
(73) Owners :
  • THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA (United States of America)
(71) Applicants :
  • THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA (United States of America)
(74) Agent: NORTON ROSE FULBRIGHT CANADA LLP/S.E.N.C.R.L., S.R.L.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2015-10-14
(87) Open to Public Inspection: 2016-04-21
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2015/055462
(87) International Publication Number: WO2016/061182
(85) National Entry: 2017-04-13

(30) Application Priority Data:
Application No. Country/Territory Date
62/063,828 United States of America 2014-10-14
62/065,973 United States of America 2014-10-20

Abstracts

English Abstract

The present invention provides methods of treating anal or vaginal tumors and cancers, comprising the step of administering to a subject a combination therapy comprising a chemo-radiation therapy and a recombinant Listeria strain. In one aspect, the present invention relates to a method of treating an anal or vaginal tumor or anal or vaginal cancer in a human subject, the method comprising the step of administering to said subject a combination therapy comprising a chemo-radiation therapy and a recombinant Listeria strain, said Listeria strain comprising a recombinant nucleic acid, said nucleic acid comprising a first open reading frame encoding a recombinant polypeptide comprising an N-terminal fragment of an LLO protein fused to a heterologous antigen or fragment thereof, thereby treating said anal or vaginal tumor or anal or vaginal cancer in said human subject.


French Abstract

La présente invention concerne des procédés de traitement de tumeurs et de cancers anaux ou vaginaux, comprenant l'étape consistant à administrer à un sujet une polythérapie comprenant une radiochimiothérapie et une souche recombinante de la Listeria. Dans un aspect, la présente invention concerne un procédé de traitement d'une tumeur anale ou vaginale ou d'un cancer anal ou vaginal chez un sujet humain, le procédé comprenant l'étape consistant à administrer audit sujet une combinaison thérapie comprenant une radiochimiothérapie et une souche recombinante de la Listeria, ladite souche recombinante de la Listeria comprenant un acide nucléique recombinant, ledit acide nucléique comprenant un premier cadre de lecture ouvert codant pour un polypeptide recombinant comprenant un fragment N-terminal d'une protéine LLO fusionné à un antigène hétérologue ou un de ses fragments, ce qui permet de traiter ladite tumeur anale ou vaginale ou ledit cancer anal ou vaginal chez ledit sujet humain.

Claims

Note: Claims are shown in the official language in which they were submitted.


What is claimed:
1. A method of treating an anal or vaginal tumor or anal or vaginal cancer
in a human
subject, the method comprising the step of administering to said subject a
combination
therapy comprising a chemo-radiation therapy and a recombinant Listeria
strain, said
Listeria strain comprising a recombinant nucleic acid, said nucleic acid
comprising a
first open reading frame encoding a recombinant polypeptide comprising an N-
terminal
fragment of an LLO protein fused to a heterologous antigen or fragment
thereof, thereby
treating said anal or vaginal tumor or anal or vaginal cancer in said human
subject.
2. The method of claim 1, wherein said anal tumor or anal cancer is anal
intraepithelial
cancer.
3. The method of claim 1, wherein said vaginal tumor or vaginal cancers is
vaginal
intraepithelial cancer.
4. The method of any one of claims 1-3, wherein said chemo-radiation therapy
is
administered following a first administration of said recombinant Listeria
strain.
5. The method of any one of claims 1-4, wherein said chemo-radiation therapy
is
administered prior to the administration of said recombinant Listeria strain.
6. The method of any one of claims 1-5, wherein said chemo-radiation therapy
is
administered following a first administration of said recombinant Listeria
strain and
prior to one to three booster administrations of said recombinant Listeria
strain.
7. The method of any one of claims 1-6, wherein said chemo-radiation therapy
is
administered concurrently with said recombinant Listeria strain.
8. The method of any one of claims 1-7, wherein said method comprises
administering four
doses of said recombinant Listeria.
66

9. The method of any one of claims 1-8, wherein the first dose of said
recombinant Listeria
is administered prior to chemo-radiation therapy and the 2th1-4th doses are
administered
every 28 days after completion of radiation.
10. The method of any one of claims 1-9, wherein the first dose of said
recombinant Listeria
is administered before chemo-radiation therapy, wherein the second dose of
ADXS-HPV
is administered during chemo-radiation therapy, and wherein the 3rd ¨ 4th
doses are
administered every 28 days following the completion of chemo-radiation
therapy.
11. The method of any one of claims 1-10, wherein said chemo-radiation
therapy comprises
mitomycin and fluorouracil (5-FU) and radiation therapy.
12. The method of any one of claims 1-11, wherein said chemo-radiation
therapy comprises
administering 2 courses of mitomycin, 5-FU with concurrent radiation (54 Gy in
30
fractions by intensity modulated radiation therapy).
13. The method of any one of claims 1-12, wherein said radiation therapy lasts
about 6
weeks.
14. The method of any one of claims 1-13, wherein said Listeria comprises a
mutation or
deletion in the endogenous prfA gene.
15. The method of any one of claims 1-14, wherein said recombinant nucleic
acid further
comprises a second open reading frame encoding a mutant prfA gene, thereby
inducing
an immune response against said anal or vaginal tumor or anal or vaginal
cancer.
16. The method of any one of claims 1-15, wherein said mutant prfA gene
encodes a PrfA
protein comprising a D133V mutation.
67

17. The recombinant Listeria of any one of claims 1-16, wherein said mutant
PrfA protein
complements a prfA genomic mutation or deletion.
18. The method of any one of claims 1-17, wherein said administering is
intravenous or oral
administering.
19. The method of any one of claims 1-18, wherein said N-terminal fragment of
an LLO
protein comprises SEQ ID NO: 2.
20. The method of any one of claims 1-19, wherein said recombinant Listeria
strain is
administered to said human subject at a dose of 1 x 10 9 - 3.31 x 10 10
organisms.
21. The method of any one of claims 1-20, wherein said recombinant Listeria
strain is a
recombinant Listeria monocytogenes strain
22. The method of any one of claims 1-21, wherein said recombinant Listeria
strain has
been passaged through an animal host, prior to the step of administering.
23. The method of any one of claims 1-22, wherein said recombinant polypeptide
is
expressed by said recombinant Listeria strain.
24. The method of any one of claim 1-23, wherein said recombinant Listeria
strain
comprises a plasmid that encodes said recombinant polypeptide.
25. The method of any one of claims 1-24, further comprising the step of
boosting said
human subject with said recombinant Listeria strain.
26. The method of any one of claims 1-25, further comprising the step of
inoculating said
human subject with an immunogenic composition that comprises or directs
expression of
said E7 antigen.
68

27. The method of any one of claims 1-26, wherein said recombinant Listeria
strain has
been stored in a frozen or lyophilized cell bank.
28. A method for inducing an anti-tumor cytotoxic T cell response in a human
subject,
comprising the step of administering to said subject the combination therapy
of any one
of claims 1-27.
29. A method of treating a human subject against a tumor or cancer, comprising
the step of
administering to said subject the recombinant Listeria strain of any one of
claims 1-28.
30. The method of any one of claims 1-29, wherein said administering is
intravenous or oral
administering.
31. The method of any one of claims 1-30, wherein said immune response
comprises
increasing a level of interferon-gamma producing cells.
32. The method of any one of claims 1-31, wherein said immune response
comprises
increasing a level of TNF-alpha producing cells.
33. The method of any one of claims 1-32, wherein said immune response
comprises an
increase of tumor infiltration by T effector cells.
34. The method of any one of claims 1-33, wherein said T effector cells are
CD8+T cells or
CD4+T cells.
35. The method of any one of claims 1-34, wherein said immune response
further comprises
epitope spreading.
36. The method of any one of claims 1-35, wherein said immune response
further comprises
induction of broad-based response to self-derived tumor antigens.
69

37. The method of any one of claims 1-36, wherein said immune response
further comprises
improvement of the overall balance of suppressor and effector immune cells in
the tumor
microenvironment.
38. The method of any one of claims 1-37, wherein said immune response
further comprises
improvement in the systemic balance of suppressor and effector immunocytes.
39. A method of treating an anal or vaginal neoplasia in a human subject, the
method
comprising the step of administering to said subject a combination therapy
comprising a
chemo-radiation therapy and a recombinant Listeria strain, said Listeria
strain
comprising a recombinant nucleic acid, said nucleic acid comprising a first
open reading
frame encoding a recombinant polypeptide comprising an N-terminal fragment of
an
LLO protein fused to a heterologous antigen or fragment thereof, thereby
treating said
anal or vaginal neoplasia in said human subject.
40. The method of claim 39, wherein said anal neoplasia is anal
intraepithelial neoplasia.
41. The method of claim 40, wherein said vaginal neoplasia is vaginal
intraepithelial
neoplasia.
42. The method of any one of claims 39-41, wherein said chemo-radiation
therapy is
administered following a first administration of said recombinant Listeria
strain.
43. The method of any one of claims 39-42, wherein said chemo-radiation
therapy is
administered prior to the administration of said recombinant Listeria strain.
44. The method of any one of claims 39-43, wherein said chemo-radiation
therapy is
administered following a first administration of said recombinant Listeria
strain and
prior to one to three booster administrations of said recombinant Listeria
strain.

45. The method of any one of claims 39-44, wherein said chemo-radiation
therapy is
administered concurrently with said recombinant Listeria strain.
46. The method of any one of claims 39-45, wherein said method comprises
administering
four doses of said recombinant Listeria.
47. The method of any one of claims 39-46, wherein the first dose of said
recombinant
Listeria is administered prior to chemo-radiation therapy and the 2nd-4th
doses are
administered every 28 days after completion of radiation.
48. The method of any one of claims 39-47, wherein the first dose of said
recombinant
Listeria is administered before chemo-radiation therapy, wherein the second
dose of
ADXS-HPV is administered during chemo-radiation therapy, and wherein the 3rd ¨
4th
doses are administered every 28 days following the completion of chemo-
radiation
therapy.
49. The method of any one of claims 39-48, wherein said chemo-radiation
therapy
comprises mitomycin and fluorouracil (5-FU) and radiation therapy.
50. The method of any one of claims 39-49, wherein said chemo-radiation
therapy
comprises administering 2 courses of mitomycin, 5-FU with concurrent radiation
(54 Gy
in 30 fractions by intensity modulated radiation therapy).
51. The method of any one of claims 39-50, wherein said radiation therapy
lasts about 6
weeks.
52. The method of any one of claims 39-51, wherein said Listeria comprises a
mutation or
deletion in the endogenous prfA gene.
71

53. The method of any one of claims 39-52, wherein said recombinant nucleic
acid further
comprises a second open reading frame encoding a mutant prfA gene, thereby
inducing
an immune response against said anal or vaginal neoplasia.
54. The method of any one of claims 39-53, wherein said mutant prfA gene
encodes a
mutant PrfA protein that comprises a D133V mutation.
55. The recombinant Listeria of any one of claims 39-54, wherein said
mutant PrfA protein
complements a prfA genomic mutation or deletion.
56. The method of any one of claims 39-55, wherein said administering is
intravenous or
oral administering.
57. The method of any one of claims 39-56, wherein said N-terminal fragment of
an LLO
protein comprises SEQ ID NO: 2.
58. The method of any one of claims 39-57, wherein said recombinant Listeria
strain is
administered to said human subject at a dose of 1 x 10 9 - 3.31 x 10 10
organisms.
59. The method of any one of claims 39-58, wherein said recombinant Listeria
strain is a
recombinant Listeria monocytogenes strain
60. The method of any one of claims 39-59, wherein said recombinant Listeria
strain has
been passaged through an animal host, prior to the step of administering.
61. The method of any one of claims 39-60, wherein said recombinant
polypeptide is
expressed by said recombinant Listeria strain.
62. The method of any one of claim 39-61, wherein said recombinant Listeria
strain
comprises a plasmid that encodes said recombinant polypeptide.
72

63. The method of any one of claims 39-62, further comprising the step of
boosting said
human subject with said recombinant Listeria strain.
64. The method of any one of claims 39-63, further comprising the step of
inoculating said
human subject with an immunogenic composition that comprises or directs
expression of
said E7 antigen.
65. The method of any one of claims 39-64, wherein said recombinant Listeria
strain has
been stored in a frozen or lyophilized cell bank.
66. A method for inducing an anti-neoplasia cytotoxic T cell response in a
human subject,
comprising the step of administering to said subject the combination therapy
of any one
of claims 39-65.
67. A method of treating a human subject against a anal or vaginal
neoplasia, comprising the
step of administering to said subject the recombinant Listeria strain of any
one of claims
39-66.
68. The method of claim any one of claims 39-67, wherein said administering
is intravenous
or oral administering.
69. The method of any one of claims 39-68, wherein said immune response
comprises
increasing a level of interferon-gamma producing cells.
70. The method of any one of claims 39-69, wherein said immune response
comprises
increasing a level of TNF-alpha producing cells.
71. The method of any one of claims 39-70, wherein said immune response
comprises an
increase of neoplasia infiltration by T effector cells.
73

72. The method of any one of claims 39-71, wherein said T effector cells are
CD8+T cells or
CD4+T cells.
73. The method of any one of claims 39-72, wherein said immune response
further
comprises epitope spreading.
74. The method of any one of claims 39-73, wherein said immune response
further
comprises induction of broad-based response to self-derived neoplasia
antigens.
75. The method of any one of claims 39-74, wherein said immune response
further
comprises improvement of the overall balance of suppressor and effector immune
cells
in the neoplasia microenvironment.
76. The method of any one of claims 39-75, wherein said immune response
further
comprises improvement in the systemic balance of suppressor and effector
immunocytes.
77. Use of a composition comprising a recombinant Listeria strain, said
Listeria strain
comprising a recombinant nucleic acid, said nucleic acid comprising a first
open reading
frame encoding a recombinant polypeptide comprising an N-terminal fragment of
an
LLO protein fused to a heterologous antigen or fragment thereof for treating
an anal or
vaginal tumor or cancer in a human subject, the treatment further comprising
the step of
administering to said subject a chemo-radiation therapy, thereby treating said
anal or
vaginal tumor or anal or vaginal cancer in said human subject.
78. The use according to claim 77, wherein said chemo-radiation therapy is
administered
following a first administration of said recombinant Listeria strain.
79. The use according to any one of claims 77-78, wherein said chemo-
radiation therapy is
administered prior to the administration of said recombinant Listeria strain.
74

80. The use according to any one of claims 77-79, wherein said chemo-
radiation therapy is
administered following a first administration of said recombinant Listeria
strain and
prior to one to three booster administrations of said recombinant Listeria
strain.
81. The use according to any one of claims 77-80, wherein said chemo-
radiation therapy is
administered concurrently with said recombinant Listeria strain.
82. The use according to any one of claims 77-81, wherein said method
comprises
administering four doses of said recombinant Listeria.
83. The use according to any one of claims 77-82, wherein the first dose of
said recombinant
Listeria is administered prior to chemo-radiation therapy and the 2nd-4th
doses are
administered every 28 days after completion of radiation.
84. The use according to any one of claims 77-83, wherein the first dose of
said recombinant
Listeria is administered before chemo-radiation therapy, wherein the second
dose of
ADXS-HPV is administered during chemo-radiation therapy, and wherein the 3rd ¨
4th
doses are administered every 28 days following the completion of chemo-
radiation
therapy.
85. The use according to any one of claims 77-84, wherein said chemo-radiation
therapy
comprises mitomycin and fluorouracil (5-FU) and radiation therapy.
86. The use according to any one of claims 77-85, wherein said chemo-radiation
therapy
comprises administering 2 courses of mitomycin, 5-FU with concurrent radiation
(54 Gy
in 30 fractions by intensity modulated radiation therapy).
87. The use according to any one of claims 77-86, wherein said radiation
therapy lasts about
6 weeks.

88. The use according to of any one of claims 77-87, wherein said Listeria
comprises a
mutation or deletion in the endogenous prfA gene.
89. The use according to any one of claims 77-88, wherein said recombinant
nucleic acid
further comprises a second open reading frame encoding a mutant prfA gene,
thereby
inducing an immune response against said anal or vaginal tumor or anal or
vaginal
cancer.
90. The use according to any one of claims 77-89, wherein said mutant prfA
gene encodes a
mutant PrfA protein that comprises a D133V mutation.
91. The recombinant Listeria of any one of claims 77-90, wherein said
mutant PrfA protein
complements a prfA genomic mutation or deletion.
92. The use according to any one of claims 77-91, wherein said
administering is intravenous
or oral administering.
93. The use according to any one of claims 77-92, wherein said N-terminal
fragment of an
LLO protein comprises SEQ ID NO: 2.
94. The use according to any one of claims 77-93, wherein said recombinant
Listeria strain
is administered to said human subject at a dose of 1 x 10 9 - 3.31 x 10 10
organisms.
95. The use according to any one of claims 77-94, wherein said recombinant
Listeria strain
is a recombinant Listeria monocytogenes strain
96. The use according to any one of claims 77-95, wherein said recombinant
Listeria strain
has been passaged through an animal host, prior to the step of administering.
97. The use according to any one of claims 77-96, wherein said recombinant
polypeptide is
expressed by said recombinant Listeria strain.
76

98. The use according to any one of claim 77-97, wherein said recombinant
Listeria strain
comprises a plasmid that encodes said recombinant polypeptide.
99. The use according to any one of claims 77-98, further comprising the step
of boosting
said human subject with said recombinant Listeria strain.
100. The use according to any one of claims 77-99, further comprising the step
of inoculating
said human subject with an immunogenic composition that comprises or directs
expression of said E7 antigen.
101. The use according to any one of claims 77-100, wherein said recombinant
Listeria strain
has been stored in a frozen or lyophilized cell bank.
102. A method for inducing an anti-tumor cytotoxic T cell response in a human
subject,
comprising the step of administering to said subject the combination therapy
of any one
of claims 77-101.
103. The use according to any one of claims 77-102, wherein treating a human
subject against
a tumor or cancer comprises the step of administering to said subject said
recombinant
Listeria strain.
104. The use according to claim any one of claims 77-103, wherein said
administering is
intravenous or oral administering.
105. The use according to any one of claims 77-104, wherein said immune
response
comprises increasing a level of interferon-gamma producing cells.
106. The use according to any one of claims 77-105, wherein said immune
response
comprises increasing a level of TNF-alpha producing cells.
77

107. The use according to any one of claims 77-106, wherein said immune
response
comprises an increase of tumor infiltration by T effector cells.
108. The use according to any one of claims 77-107, wherein said T effector
cells are CD8+T
cells or CD4+T cells.
109. The use according to any one of claims 77-108, wherein said immune
response further
comprises epitope spreading.
110. The use according to any one of claims 77-109, wherein said immune
response further
comprises induction of broad-based response to self-derived tumor antigens.
111. The use according to any one of claims 77-110, wherein said immune
response further
comprises improvement of the overall balance of suppressor and effector immune
cells
in the tumor microenvironment.
112. The use according to any one of claims 77-111, wherein said immune
response further
comprises improvement in the systemic balance of suppressor and effector
immunocytes.
113. Use of a composition comprising a recombinant Listeria strain, said
Listeria strain
comprising a recombinant nucleic acid, said nucleic acid comprising a first
open reading
frame encoding a recombinant polypeptide comprising an N-terminal fragment of
an
LLO protein fused to a heterologous antigen or fragment thereof for treating
an anal or
vaginal neoplasia in a human subject, the treatment further comprising the
step of
administering to said subject a chemo-radiation therapy, thereby treating said
anal or
vaginal neoplasia in said human subject.
114. The use according to claim 113, wherein said anal neoplasia is anal
intraepithelial
neoplasia.
78

115. The use according to claim 113, wherein said vaginal neoplasia is vaginal
intraepithelial
neoplasia.
116. The use according to any one of claims 113-115, wherein said chemo-
radiation therapy
is administered following a first administration of said recombinant Listeria
strain.
117. The use according to any one of claims 113-116, wherein said chemo-
radiation therapy
is administered prior to the administration of said recombinant Listeria
strain.
118. The use according to any one of claims 113-117, wherein said chemo-
radiation therapy
is administered following a first administration of said recombinant Listeria
strain and
prior to one to three booster administrations of said recombinant Listeria
strain.
119. The use according to any one of claims 113-118, wherein said chemo-
radiation therapy
is administered concurrently with said recombinant Listeria strain.
120. The use according to any one of claims 113-119, wherein said method
comprises
administering four doses of said recombinant Listeria.
121. The use according to any one of claims 113-120, wherein the first dose of
said
recombinant Listeria is administered prior to chemo-radiation therapy and the
2nd-4th
doses are administered every 28 days after completion of radiation.
122. The use according to any one of claims 113-121, wherein the first dose of
said
recombinant Listeria is administered before chemo-radiation therapy, wherein
the
second dose of ADXS-HPV is administered during chemo-radiation therapy, and
wherein the 3rd ¨ 4th doses are administered every 28 days following the
completion of
chemo-radiation therapy.

79

123. The use according to any one of claims 113-122, wherein said chemo-
radiation therapy
comprises mitomycin and fluorouracil (5-FU) and radiation therapy.
124. The use according to any one of claims 113-123, wherein said chemo-
radiation therapy
comprises administering 2 courses of mitomycin, 5-FU with concurrent radiation
(54 Gy
in 30 fractions by intensity modulated radiation therapy).
125. The use according to any one of claims 113-124, wherein said radiation
therapy lasts
about 6 weeks.
126. The use according to any one of claims 113-125, wherein said Listeria
comprises a
mutation or deletion in the endogenous pr.function.A gene.
127. The use according to any one of claims 113-126, wherein said recombinant
nucleic acid
further comprises a second open reading frame encoding a mutant pr.function.A
gene, thereby
inducing an immune response against said anal or vaginal neoplasia.
128. The use according to any one of claims 113-127, wherein said mutant
pr.function.A gene encodes
a mutant Pr.function.A protein that comprises a D133V mutation.
129. The recombinant Listeria of claim 113-128, wherein said mutant
Pr.function.A protein
complements a pr.function.A genomic mutation or deletion.
130. The use according to any one of claims 113-129, wherein said
administering is
intravenous or oral administering.
131. The use according to any one of claims 113-130, wherein said N-terminal
fragment of an
LLO protein comprises SEQ ID NO: 2.
132. The use according to any one of claims 113-131, wherein said recombinant
Listeria
strain is administered to said human subject at a dose of 1 x 10 9 - 3.31 x 10
10 organisms.


133. The use according to any one of claims 113-132, wherein said recombinant
Listeria
strain is a recombinant Listeria monocytogenes strain
134. The use according to any one of claims 113-133, wherein said recombinant
Listeria
strain has been passaged through an animal host, prior to the step of
administering.
135. The use according to any one of claims 113-134, wherein said recombinant
polypeptide
is expressed by said recombinant Listeria strain.
136. The use according to any one of claim 113-135, wherein said recombinant
Listeria strain
comprises a plasmid that encodes said recombinant polypeptide.
137. The use according to any one of claims 113-136, further comprising the
step of boosting
said human subject with said recombinant Listeria strain.
138. The use according to any one of claims 113-137, further comprising the
step of
inoculating said human subject with an immunogenic composition that comprises
or
directs expression of said E7 antigen.
139. The use according to any one of claims 113-138, wherein said recombinant
Listeria
strain has been stored in a frozen or lyophilized cell bank.
140. A method for inducing an anti-neoplasia cytotoxic T cell response in a
human subject,
comprising the step of administering to said subject the combination therapy
of any one
of claims 113-139.
141. The use according to any one of claims 113-140, wherein treating a human
subject
against an anal or vaginal neoplasia comprises the step of administering to
said subject
said recombinant Listeria strain.

81

142. The use according to any one of claims 113-141, wherein said
administering is
intravenous or oral administering.
143. The use according to any one of claims 113-142, wherein said immune
response
comprises increasing a level of interferon-gamma producing cells.
144. The use according to any one of claims 113-143, wherein said immune
response
comprises increasing a level of TNF-alpha producing cells.
145. The use according to any one of claims 113-144, wherein said immune
response
comprises an increase of neoplasia infiltration by T effector cells.
146. The use according to any one of claims 113-145, wherein said T effector
cells are
CD8+T cells or CD4+T cells.
147. The use according to any one of claims 113-146, wherein said immune
response further
comprises epitope spreading.
148. The use according to any one of claims 113-147, wherein said immune
response further
comprises induction of broad-based response to self-derived neoplasia
antigens.
149. The use according to any one of claims 113-148, wherein said immune
response further
comprises improvement of the overall balance of suppressor and effector immune
cells
in the neoplasia microenvironment.
150. The use according to any one of claims 113-149, wherein said immune
response further
comprises improvement in the systemic balance of suppressor and effector
immunocytes.

82

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
COMBINATION THERAPY FOR USE IN CANCER THERAPY
FIELD OF INVENTION
[001] The present invention provides methods of treating anal or vaginal
tumors and anal or
vaginal cancers, comprising the step of administering to a subject a
combination therapy
comprising a chemo-radiation therapy and a recombinant Listeria strain.
BACKGROUND OF THE INVENTION
[002] Listeria monocytogenes (Lm) is a food-borne gram-positive bacterium that
can
occasionally cause disease in humans, in particular elderly individuals,
newborns, pregnant
women and immunocompromised individuals. In addition to strongly activating
innate
immunity and inducing a cytokine response that enhances antigen-presenting
cell (APC)
function, Lm has the ability to replicate in the cytosol of APCs after
escaping from the
phagolysosome, mainly through the action of the listeriolysin 0 (LLO) protein.
This unique
intracellular life cycle allows antigens secreted by Lm to be processed and
presented in the
context of both MHC class I and II molecules, resulting in potent cytotoxic
CD8+ and Thl
CD4+ T-cell¨mediated immune responses. Lm has been extensively investigated as
a vector
for cancer immunotherapy in pre-clinical models.
[003] Persistent infection with high-oncogenic risk human papillomavirus (HR-
HPV) types
is recognized as a necessary, but not sufficient, cause of invasive carcinoma
of the cervix
(ICC). HPVs 16 and 18 are the most prevalent types in malignant lesions,
accounting for over
70% of ICC and over 50% of high-grade precursor lesions.
[004] Anal cancer is a rare malignancy that begins in the anus, which is the
opening at the
end of the rectum. The American Cancer Society estimates that 7,270 cases of
anal cancer
will be diagnosed in 2015 (with the incidence still increasing) and about
1,010 deaths will
occur that year from anal cancer. Approximately half of all anal cancers are
diagnosed before
the malignancy has spread beyond the primary site, whereas 13% to 25% are
diagnosed after
the cancer has spread to the lymph nodes, and 10% are diagnosed after the
cancer has spread
to distant organs, or has metastasized. When it is found early, anal cancer is
highly treatable,
however, if the cancer has spread to distant organs, about one in five
patients lives for five
years or more.
1

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[005] Receptive anal intercourse is strongly related to the development of
anal cancer. Anal
infection with human papillomavirus (HPV) resulting in genital warts is a
major risk factor
for the cancer, and immunocompromised patients, are prone to get anal cancer.
In this
subgroup, the prognosis is worse, than for non-immunocompromised patients.
[006] Anal cancer is primarily treated with a combination of chemotherapy and
radiation.
This reduces the need for a colostomy and carries a 5-year survival rate of
over 70%. Despite
this, the treatment of anal cancer, in this fashion, has not changed since
1974 and surgery is
reserved only for patients failing the above therapy. Hence, there exists a
need for alternative
approaches to treating anal cancer. One such approach is an immunotherapeutic
approach
using recombinant attenuated live vaccine vectors, such as a Listeria
monocytogenes vaccine
vector. The present invention provides an attenuated live Listeria vaccine
vector for treating
anal cancer.
SUMMARY OF THE INVENTION
[007] In one aspect, the present invention relates to a method of treating an
anal or vaginal
tumor or anal or vaginal cancer in a human subject, the method comprising the
step of
administering to said subject a combination therapy comprising a chemo-
radiation therapy
and a recombinant Listeria strain, said Listeria strain comprising a
recombinant nucleic acid,
said nucleic acid comprising a first open reading frame encoding a recombinant
polypeptide
comprising an N-terminal fragment of an LLO protein fused to a heterologous
antigen or
fragment thereof, thereby treating said anal or vaginal tumor or anal or
vaginal cancer in said
human subject.
[008] In another aspect, the present invention relates to a method of treating
an anal or
vaginal neoplasia in a human subject, the method comprising the step of
administering to said
subject a combination therapy comprising a chemo-radiation therapy and a
recombinant
Listeria strain, said Listeria strain comprising a recombinant nucleic acid,
said nucleic acid
comprising a first open reading frame encoding a recombinant polypeptide
comprising an N-
terminal fragment of an LLO protein fused to a heterologous antigen or
fragment thereof,
thereby treating said anal or vaginal neoplasia in said human subject.
[009] In a further aspect, the present invention relates to the use of a
composition
comprising a recombinant Listeria strain, said Listeria strain comprising a
recombinant
nucleic acid, said nucleic acid comprising a first open reading frame encoding
a recombinant
polypeptide comprising an N-terminal fragment of an LLO protein fused to a
heterologous
2

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
antigen or fragment thereof for treating an anal or vaginal tumor or anal or
vaginal cancer in a
human subject, the treatment further comprising the step of administering to
said subject a
chemo-radiation therapy, thereby treating said anal or vaginal tumor or anal
or vaginal cancer
in said human subject.
[0010] In a yet further aspect, the present invention relates to the use of a
composition
comprising a recombinant Listeria strain, said Listeria strain comprising a
recombinant
nucleic acid, said nucleic acid comprising a first open reading frame encoding
a recombinant
polypeptide comprising an N-terminal fragment of an LLO protein fused to a
heterologous
antigen or fragment thereof for treating an anal or vaginal neoplasia in a
human subject, the
treatment further comprising the step of administering to said subject a chemo-
radiation
therapy, thereby treating said anal or vaginal neoplasia in said human
subject.
[0011] In a related aspect, the present invention relates to a method of
eliciting an anti-tumor
cytotoxic T cell response in a human subject comprising administering to said
subject said
combination therapy.
[0012] Other features and advantages disclosed herein will become apparent
from the
following detailed description examples and figures. It should be understood,
however, that
the detailed description and the specific examples while indicating preferred
embodiments of
the invention are given by way of illustration only, since various changes and
modifications
within the spirit and scope of the invention will become apparent to those
skilled in the art
from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
[0013] The subject matter regarded as the invention is particularly pointed
out and distinctly
claimed in the concluding portion of the specification. The invention,
however, both as to
organization and method of operation, together with objects, features, and
advantages thereof,
may best be understood by reference to the following detailed description when
read with the
accompanying drawings in which:
[0014] Figure 1. Lm-E7 and Lm-LLO-E7 use different expression systems to
express and
secrete E7. Lm-E7 was generated by introducing a gene cassette into the orfZ
domain of the
L. monocytogenes genome (A). The hly promoter drives expression of the hly
signal
sequence and the first five amino acids (AA) of LLO followed by HPV-16 E7. B),
Lm-LLO-
E7 was generated by transforming the PrfA- strain XFL-7 with the plasmid pGG-
55. pGG-55
3

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
has the hly promoter driving expression of a nonhemolytic fusion of LLO-E7.
pGG-55 also
contains the PrfA gene to select for retention of the plasmid by XFL-7 in
vivo.
[0015] Figure 2. Lm-E7 and Lm-LLO-E7 secrete E7. Lm-Gag (lane 1), Lm-E7 (lane
2), Lm-
LLO-NP (lane 3), Lm-LLO-E7 (lane 4), XFL-7 (lane 5), and 10403S (lane 6) were
grown
overnight at 37 C in Luria-Bertoni broth. Equivalent numbers of bacteria, as
determined by
OD at 600 nm absorbance, were pelleted and 18 ml of each supernatant was TCA
precipitated. E7 expression was analyzed by Western blot. The blot was probed
with an anti-
E7 mAb, followed by HRP-conjugated anti-mouse (Amersham), then developed using
ECL
detection reagents.
[0016] Figure 3. Tumor immunotherapeutic efficacy of LLO-E7 fusions. Tumor
size in
millimeters in mice is shown at 7, 14, 21, 28 and 56 days post tumor-
inoculation. Naive mice:
open-circles; Lm-LLO-E7: filled circles; Lm-E7: squares; Lm-Gag: open
diamonds; and Lm-
LLO-NP: filled triangles.
[0017] Figure 4. Splenocytes from Lm-LLO-E7-immunized mice proliferate when
exposed
to TC-1 cells. C57BL/6 mice were immunized and boosted with Lm-LLO-E7, Lm-E7,
or
control rLm strains. Splenocytes were harvested 6 days after the boost and
plated with
irradiated TC-1 cells at the ratios shown. The cells were pulsed with 3H
thymidine and
harvested. Cpm is defined as (experimental cpm) - (no-TC-1 control).
[0018] Figure 5 A. Induction of E7-specific IFN-gamma-secreting CD8+ T cells
in the
spleens and the numbers penetrating the tumors, in mice administered TC-1
tumor cells and
subsequently administered Lm-E7, Lm-LLO-E7, Lm-ActA-E7, or no vaccine (naive).
Figure
5 B. Induction and penetration of E7 specific CD8+ cells in the spleens and
tumors of the mice
described for (A).
[0019] Figure 6. Listeria constructs containing PEST regions induce a higher
percentage of
E7-specific lymphocytes within the tumor. Figure 6 A. representative data from
1 experiment.
Figure 6 B. average and SE of data from all 3 experiments.
[0020] Figure 7A. Effect of passaging on bacterial load (virulence) of
recombinant Listeria
vaccine vectors. Top panel. Lm-Gag. Bottom panel. Lm-LLO-E7. Figure 7B. Effect
of
passaging on bacterial load of recombinant Lm-E7 in the spleen. Average CFU of
live
bacteria per milliliter of spleen homogenate from four mice is depicted.
4

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0021] Figure 8 shows induction of antigen-specific CD8+ T-cells for HIV-Gag
and LLO
after administration of passaged Lm-Gag versus unpassaged Lm-Gag. Mice were
immunized
with 103 (A, B, E, F) or 105 (C, D, G, H) CFU passaged Listeria vaccine
vectors, and antigen-
specific T-cells were analyzed. B, D, F, H: unpassaged Listeria vaccine
vectors. A-D immune
response to MHC class I HIV-Gag peptide. E-H: immune response to an LLO
peptide. I:
splenocytes from mice immunized with 105 CFU passaged Lm-Gag stimulated with a
control
peptide from HPV E7.
[0022] Figure 9A shows plasmid isolation throughout LB stability study. Figure
9B shows
plasmid isolation throughout TB stability study. Figure 9C shows quantitation
of TB stability
study.
[0023] Figure 10 shows numbers of viable bacteria chloramphenicol (CAP)-
resistant and
CAP-sensitive colony-forming units (CFU) from bacteria grown in LB. Dark bars:
CAP;
white bars: CAP-. The two dark bars and two white bars for each time point
represent
duplicate samples.
[0024] Figure 11 shows numbers of viable bacteria CAP-resistant and CAP-
sensitive CFU
from bacteria grown in TB. Dark bars: CAP; white bars: CAP. The two dark bars
and two
white bars for each time point represent duplicate samples.
[0025] Figure 12. Actual chromatograms showing the region of the D133V
mutation
(arrows). The mixture ratio is shown in parentheses.
[0026] Figure 13. Representation of the location of the ADV451, 452 and 453
primers and
the segment of the PrfA gene amplified in the reaction.
[0027] Figure 14. Specificity of the PCR reaction using primers ADV451 and
ADV453.
[0028] Figure 15. Specificity of the PCR reaction using primers ADV452 and
ADV453.
[0029] Figure 16. Sensitivity of the PCR reaction to detect the wild-type PrfA
sequence using
the primer ADV452 and 1 ng as the initial amount of DNA.
[0030] Figure 17. Sensitivity of the PCR reaction to detect the wild-type PrfA
sequence using
the primer ADV452 and 5 ng as the initial amount of DNA.
[0031] Figure 18. Average density of the bands from the PCR depicted in figure
16.
5

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0032] Figure 19. Average density of the bands from the PCR depicted in figure
17.
[0033] Figure 20. Validation of the PCR reaction to detect the wild-type PrfA
sequence using
the primer ADV452.
[0034] Figure 21. Average density of the bands from the PCR depicted in figure
16.
[0035] Figure 22. Analysis of the D133V PrfA mutation in the Lm-LLO-E7. A,
Original
image used for densitometry; B, Image was digitally enhanced to facilitate the
visualization of
the low density bands.
[0036] Figure 23. Shows treatment schedule #1 of the patient. The schedule
comprises
administering a first dose of ADXS-HPV (ADX) before chemo-radiation and the 2-
4th doses
are given every 28 days after completion of radiation.
[0037] Figure 24. Shows treatment schedule #2 of the patient where the second
dose of
ADXS-HPV is administered during chemo-radiation.
[0038] It will be appreciated that for simplicity and clarity of illustration,
elements shown in
the Figs. have not necessarily been drawn to scale. For example, the
dimensions of some of
the elements may be exaggerated relative to other elements for clarity.
Further, where
considered appropriate, reference numerals may be repeated among the Figs. to
indicate
corresponding or analogous elements.
DETAILED DESCRIPTION OF THE INVENTION
[0039] The present invention disclose, in some embodiments, methods of
treating, protecting
against, and inducing an immune response against a disease, comprising the
step of
administering to a subject a recombinant Listeria strain, expressing a fusion
peptide
comprising a listeriolysin 0 (LLO) fragment and a heterologous antigen
expressed by said
disease or fragment thereof. The present invention also provides methods for
inducing an anti-
disease cytotoxic T-cell (CTL) response in a human subject and treating
disorders, and
symptoms associated with said disease comprising administration of the
recombinant Listeria
strain. In one embodiment, provided herein is a recombinant Listeria strain,
said recombinant
Listeria strain comprising a recombinant nucleic acid, said nucleic acid
comprising a first
open reading frame encoding a recombinant polypeptide comprising a first an N-
terminal
fragment of an LLO protein fused to a heterologous antigen or fragment
thereof, and wherein
said recombinant nucleic acid further comprises a second open reading frame
encoding a
6

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
mutant prfA gene. In one embodiment, the mutant PifA gene is one that encodes
a point
mutation from amino acid D or Asp or Aspartate (or Aspartic acid) to amino
acid V or Val or
Valine at the 133rd amino acid position. In one embodiment, a recombinant
Listeria strain
disclosed herein comprises a prfA mutation or deletion that is complemented
via a plasmid
comprised by the same Listeria, wherein the plasmid comprises a mutant prfA
gene encoding
a mutant PrfA protein comprising a D133V amino acid substitution.
[0040] In another embodiment, the recombinant Listeria is an attenuated
Listeria.
"Attenuation" and "attenuated" may encompass a bacterium, virus, parasite,
infectious
organism, prion, tumor cell, gene in the infectious organism, and the like,
that is modified to
reduce toxicity to a host. The host can be a human or animal host, or an
organ, tissue, or cell.
The bacterium, to give a non-limiting example, can be attenuated to reduce
binding to a host
cell, to reduce spread from one host cell to another host cell, to reduce
extracellular growth, or
to reduce intracellular growth in a host cell. Attenuation can be assessed by
measuring, e.g.,
an indicum or indicia of toxicity, the LID, the rate of clearance from an
organ, or the
competitive index (see, e.g., Auerbuch, et al. (2001) Infect. Immunity 69:5953-
5957).
Generally, an attenuation results an increase in the LD,oand/or an increase in
the rate of
clearance by at least 25%; more generally by at least 50%; most generally by
at least 100%
(2-fold); normally by at least 5-fold; more normally by at least 10-fold; most
normally by at
least 50-fold; often by at least 100-fold; more often by at least 500-fold;
and most often by at
least 1000-fold; usually by at least 5000-fold; more usually by at least
10,000-fold; and most
usually by at least 50,000-fold; and most often by at least 100,000-fold.
[0041] It will be well appreciated by a skilled artisan that the term
"Attenuated gene" may
encompass a gene that mediates toxicity, pathology, or virulence, to a host,
growth within the
host, or survival within the host, where the gene is mutated in a way that
mitigates, reduces,
or eliminates the toxicity, pathology, or virulence. The reduction or
elimination can be
assessed by comparing the virulence or toxicity mediated by the mutated gene
with that
mediated by the non-mutated (or parent) gene. "Mutated gene" encompasses
deletions, point
mutations, and frameshift mutations in regulatory regions of the gene, coding
regions of the
gene, non-coding regions of the gene, or any combination thereof. In one
embodiment,
provided herein is a method of treating an anal or vaginal tumor or anal or
vaginal cancer in a
human subject, the method comprising the step of administering to said subject
a combination
therapy comprising a chemo-radiation therapy and a recombinant Listeria
strain, said Listeria
strain comprising a recombinant nucleic acid, said nucleic acid comprising a
first open
7

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
reading frame encoding a recombinant polypeptide comprising an N-terminal
fragment of an
LLO protein fused to a heterologous antigen or fragment thereof, thereby
treating said anal or
vaginal tumor or anal or vaginal cancer in said human subject.
[0042] In one embodiment, said chemo-radiation therapy is administered
following a first
administration of said recombinant Listeria strain. In another embodiment,
said chemo-
radiation therapy is administered prior to the administration of said
recombinant Listeria
strain. In another embodiment, said chemo-radiation therapy is administered
following a first
administration of said recombinant Listeria strain and prior to one to three
booster
administrations of said recombinant Listeria strain. In another embodiment,
said chemo-
radiation therapy is administered concurrently with said recombinant Listeria
strain.
[0043] In one embodiment, the method disclosed herein comprises administering
four doses
of a recombinant Listeria provided herein. In one embodiment the recombinant
Listeria
expresses a fusion protein of N-terminal LLO and a heterologous antigen. In
another
embodiment, the heterologous antigen is human papilloma virus E7 antigen (HPV-
E7). In
another embodiment, the HPV antigen is HPVE6.
[0044] In one embodiment, the first dose of said recombinant Listeria in the
combination
therapy provided herein is administered prior to chemo-radiation therapy and
the 2nd-4th doses
are administered every 28 days after completion of radiation (see Example 11
herein). In
another embodiment, the first dose of said recombinant Listeria is
administered before
chemo-radiation therapy, the second dose of ADXS-HPV is administered during
chemo-
radiation therapy, and the 3rd ¨ 4th doses are administered every 28 days
following the
completion of chemo-radiation therapy (see Example 11).
[0045] In one embodiment, provided herein is a chemo-radiation regiment or
chemo-
radiation therapy for use in combination with the recombinant Listeria
provided herein. In
another embodiment, the chemo-radiation therapy provided herein comprises
mitomycin and
fluorouracil (5-FU) and radiation therapy. In another embodiment, the chemo-
radiation
theraphy can comprise any other chemotherapeutic agents known in the art,
including but not
limited to, Cyclophosphamide, Mechlorethamine, Chlorambucil, Melphalan,
Nitrosoureas,
Temozolomide, Azacitidine, Azathioprine, Capecitabine, Cytarabine,
Doxifluridine,
Gemcitabine, Hydroxyurea, Mercaptopurine, Methotrexate, or Tioguanine
(formerly
Thioguanine).
8

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0046] In one embodiment, the chemo-radiation regiment or chemo-radiation
therapy
provided herein comprises administering 2 courses of mitomycin, 5-FU with
concurrent
radiation (54 Gy in 30 fractions by intensity modulated radiation therapy).
[0047] In one embodiment, the radiation provided herein lasts about 6 weeks.
In another
embodiment, the radiation lasts 3 weeks. In another embodiment, the radiation
lasts 4 weeks.
In another embodiment, the radiation lasts, 5 weeks. In another embodiment,
the radiation
lasts 7 weeks. In another embodiment, the radiation lasts 8 weeks. In another
embodiment, the
radiation lasts 6-8 weeks. In another embodiment, the radiation lasts 4-6
weeks. In another
embodiment, the radiation lasts 2-4 weeks. In another embodiment, the
radiation lasts 8-10
weeks.
[0048] In another embodiment, provided herein is a method of eliciting an anti-
tumor
cytotoxic T cell response in a human subject comprising administering to said
subject said
combination therapy.
[0049] In one embodiment, provided herein is a method for inducing an immune
response
against a tumor or a cancer in a human subject, the method comprising the step
of
administering to said subject a recombinant Listeria strain comprising a
recombinant nucleic
acid, said nucleic acid comprising a first open reading frame encoding a
recombinant
polypeptide comprising an N-terminal fragment of an LLO protein fused to a
heterologous
antigen or fragment thereof, is, wherein said recombinant nucleic acid further
comprises a
second open reading frame encoding a mutant prfA gene, thereby inducing an
immune
response against a tumor or a cancer. In one embodiment, the present invention
provides a
method of treating a cancer in a human subject, comprising the step of
administering to the
subject the recombinant Listeria strain provided herein. In another
embodiment, the present
invention provides a method of protecting a human subject against a cervical
cancer,
comprising the step of administering to the subject the recombinant Listeria
strain provided
herein. In another embodiment, the recombinant Listeria strain expresses the
recombinant
polypeptide. In another embodiment, the recombinant Listeria strain comprises
a plasmid that
encodes the recombinant polypeptide. In another embodiment, the method further
comprises
the step of boosting the human subject with a recombinant Listeria strain of
the present
invention. In another embodiment, the method further comprises the step of
boosting the
human subject with an immunogenic composition comprising a heterologous
antigen or
fragment thereof provided herein. In another embodiment, the method further
comprises the
step of boosting the human subject with an immunogenic composition that
directs a cell of the
9

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
subject to express the heterologous antigen. In another embodiment, the cell
is a tumor cell. In
another embodiment, the method further comprises the step of boosting the
human subject
with the vaccine of the present invention.
[0050] In one embodiment, the present invention provides a method of inducing
an anti-
tumor or an anti-cancer immune response in a human subject, the method
comprising the step
of administering to said subject a composition comprising a recombinant
Listeria strain
comprising a recombinant nucleic acid, said nucleic acid comprising a first
open reading
frame encoding a recombinant polypeptide comprising an N-terminal fragment of
an LLO
protein fused to a heterologous antigen or fragment thereof, wherein said
recombinant nucleic
acid further comprises a second open reading frame encoding a metabolic
enzyme, thereby
inducing an immune response against a tumor or a cancer. In another
embodiment, said
Listeria comprises a mutation in the endogenous prfA gene. In another
embodiment, the
Listeria comprises a mutation or deletion in the endogenous dal/dat and actA
genes.
[0051] In one embodiment, the nucleic acid molecule provided herein comprises
a first open
reading frame encoding recombinant polypeptide comprising a heterologous
antigen or
fragment thereof. In another embodiment, the recombinant polypeptide further
comprises a N-
terminal LLO fused to the heterologous antigen. In another embodiment, the
nucleic acid
molecule provided herein further comprises a second open reading frame
encoding a
metabolic enzyme. In another embodiment, the metabolic enzyme complements an
endogenous gene that is lacking in the chromosome of the recombinant Listeria
strain. In
another embodiment, the metabolic enzyme encoded by the second open reading
frame is an
alanine racemase enzyme (dal). In another embodiment, the metabolic enzyme
encoded by the
second open reading frame is a D-amino acid transferase enzyme (dat). In
another
embodiment, the Listeria strains provided herein comprise a mutation, a
deletion or
inactivation in the genomic dal, dat, or actA genes. In another embodiment,
the Listeria
strains provided herein comprise a mutation, a deletion or inactivation in the
genomic dal,
dat, and actA genes. In another embodiment, the Listeria lack the genomic dal,
dat or actA
genes. In another embodiment, the Listeria lack the genomic dal, dat and actA
genes.
[0052] In one embodiment, the fragment thereof in the context of LLO proteins
and ActA proteins
disclosed herein refer to a peptide or polypeptide comprising an amino acid
sequence of at least 5
contiguous amino acid residues of the LLO or ActA proteins. In another
embodiment, the term refers
to a peptide or polypeptide comprising an amino acid sequence of at least of
at least 10 contiguous
amino acid residues, at least 15 contiguous amino acid residues, at least 20
contiguous amino acid

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
residues, at least 25 contiguous amino acid residues, at least 40 contiguous
amino acid residues, at
least 50 contiguous amino acid residues, at least 60 contiguous amino
residues, at least 70 contiguous
amino acid residues, at least 80 contiguous amino acid residues, at least 90
contiguous amino acid
residues, at least 100 contiguous amino acid residues, at least 125 contiguous
amino acid residues, at
least 150 contiguous amino acid residues, at least 175 contiguous amino acid
residues, at least 200
contiguous amino acid residues, at least 250 contiguous amino acid residues of
the amino acid
sequence, at least 300 contiguous amino acid residues, at least 350 contiguous
amino acid residues of,
at least 400 contiguous amino acid residues, or at least 450 contiguous amino
acid residues of an LLO
or ActA protein or polypeptide.
[0053] In another embodiment, the fragment is a functional fragment that
elicits an immune
response against a disease-associated antigen when in the form of an N-
terminal
LLO/heterologous antigen fusion protein or N-terminal ActA/heterologous
antigen fusion
protein). In another embodiment, the fragment is an immunogenic fragment. In
another
embodiment, the fragment is functional in a non-fused form.
[0054] The present invention, in certain embodiments, provides codon
optimization of a
nucleic acid heterologous to Listeria, or of a nucleic acid endogenous to
Listeria. The optimal
codons utilized by L. monocytogenes for each amino acid are shown US Patent
Publication
2007/0207170, which is hereby incorporated by reference herein. A nucleic acid
is codon-
optimized if at least one codon in the nucleic acid is replaced with a codon
that is more
frequently used by L. monocyto genes for that amino acid than the codon in the
original
sequence.
[0055] The N-terminal LLO protein fragment and heterologous antigen are, in
another
embodiment, fused directly to one another. In another embodiment, the genes
encoding the N-
terminal LLO protein fragment and the heterologous antigen are fused directly
to one another.
In another embodiment, the N-terminal LLO protein fragment and the
heterologous antigen
are attached via a linker peptide. In another embodiment, the N-terminal LLO
protein
fragment and the heterologous antigen are attached via a heterologous peptide.
In another
embodiment, the N-terminal LLO protein fragment is N-terminal to the
heterologous antigen.
In another embodiment, the N-terminal LLO protein fragment is the N-terminal-
most portion
of the fusion protein. As disclosed herein, recombinant Listeria strains
expressing LLO-
antigen fusions induce anti-tumor immunity (Example 1), elicit antigen-
specific T cell
proliferation (Example 2), generate antigen-specific, and tumor-infiltrating T
cells (Example
3).
11

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0056] In another embodiment, the present invention provides a method of
treating a cervical
cancer in a human subject, comprising the step of administering to the subject
a recombinant
Listeria strain, the recombinant Listeria strain comprising a recombinant
polypeptide
comprising an N-terminal fragment of an LLO protein and an HPV E7 antigen,
whereby the
recombinant Listeria strain induces an immune response against the E7 antigen,
thereby
treating a cervical cancer in a human subject. In another embodiment, the
recombinant
Listeria strain expresses the recombinant polypeptide. In another embodiment,
the
recombinant Listeria strain comprises a plasmid that encodes the recombinant
polypeptide.
[0057] In another embodiment, the present invention provides a method of
protecting a
human subject against a cervical cancer, comprising the step of administering
to the subject a
recombinant Listeria strain, the recombinant Listeria strain comprising a
recombinant
polypeptide comprising an N-terminal fragment of an LLO protein and an HPV E7
antigen,
whereby the recombinant Listeria strain induces an immune response against the
E7 antigen,
thereby protecting a human subject against a cervical cancer. In another
embodiment, the
recombinant Listeria strain expresses the recombinant polypeptide. In another
embodiment,
the recombinant Listeria strain comprises a plasmid that encodes the
recombinant
polypeptide.
[0058] In another embodiment, the present invention provides a method for
inducing an
immune response against a cervical cancer in a human subject, comprising the
step of
administering to the subject a recombinant Listeria strain, the recombinant
Listeria strain
comprising a recombinant polypeptide comprising an N-terminal fragment of an
LLO protein
and an HPV E7 antigen, thereby inducing an immune response against a cervical
cancer in a
human subject. In another embodiment, the recombinant Listeria strain
expresses the
recombinant polypeptide. In another embodiment, the recombinant Listeria
strain comprises a
plasmid that encodes the recombinant polypeptide.
[0059] In another embodiment, the present invention provides a method of
treating a cervical
cancer in a human subject, comprising the step of administering to the subject
a recombinant
Listeria strain, the recombinant Listeria strain comprising a recombinant
polypeptide
comprising an N-terminal fragment of an ActA protein and heterologous antigen,
whereby the
recombinant Listeria strain induces an immune response against the
heterologous antigen,
thereby treating a cervical cancer in a human subject. In another embodiment,
the
recombinant Listeria strain expresses the recombinant polypeptide. In another
embodiment,
the recombinant Listeria strain comprises a plasmid that encodes the
recombinant
polypeptide.
12

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0060] In another embodiment, the present invention provides a method of
protecting a
human subject against a cervical cancer, comprising the step of administering
to the subject a
recombinant Listeria strain, the recombinant Listeria strain comprising a
recombinant
polypeptide comprising an N-terminal fragment of an ActA protein and a
heterologous
antigen, whereby the recombinant Listeria strain induces an immune response
against the
heterologous antigen, thereby protecting a human subject against a cervical
cancer. In another
embodiment, the recombinant Listeria strain expresses the recombinant
polypeptide. In
another embodiment, the recombinant Listeria strain comprises a plasmid that
encodes the
recombinant polypeptide.
[0061] In another embodiment, the present invention provides a method for
inducing an
immune response against a cervical cancer in a human subject, comprising the
step of
administering to the subject a recombinant Listeria strain, the recombinant
Listeria strain
comprising a recombinant polypeptide comprising an N-terminal fragment of an
heterologous
protein and a heterologous antigen, thereby inducing an immune response
against a cervical
cancer in a human subject. In another embodiment, the recombinant Listeria
strain expresses
the recombinant polypeptide. In another embodiment, the recombinant Listeria
strain
comprises a plasmid that encodes the recombinant polypeptide.
[0062] The N-terminal ActA protein fragment and the heterologous antigen are,
in another
embodiment, fused directly to one another. In another embodiment, the genes
encoding the N-
terminal ActA protein fragment and heterologous antigen are fused directly to
one another. In
another embodiment, the N-terminal ActA protein fragment and heterologous
antigen are
attached via a linker peptide. In another embodiment, the N-terminal ActA
protein fragment
and heterologous antigen are attached via a heterologous peptide. In another
embodiment, the
N-terminal ActA protein fragment is N-terminal to the heterologous antigen. In
another
embodiment, the N-terminal ActA protein fragment is the N-terminal-most
portion of the
fusion protein.
[0063] In another embodiment, the present invention provides a method of
inducing an
immune response against a cervical cancer in a human subject, comprising the
step of
administering to the subject a recombinant Listeria strain, the recombinant
Listeria strain
comprising a recombinant polypeptide comprising a PEST amino acid sequence-
containing
peptide and a heterologous antigen, whereby the recombinant Listeria strain
induces an
immune response against the heterologous antigen, thereby treating a cervical
cancer in a
human subject. In another embodiment, the recombinant Listeria strain
expresses the
recombinant polypeptide. In another embodiment, the recombinant Listeria
strain comprises a
13

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
plasmid that encodes the recombinant polypeptide. In another embodiment, the
method
protects a human subject against a cervical. In another embodiment, the method
treats a
cervical cancer in said human subject.
[0064] The PEST amino acid amino acid sequence-containing peptide and
heterologous
antigen are, in another embodiment, fused directly to one another. In another
embodiment, the
genes encoding the PEST amino acid sequence-containing peptide and
heterologous antigen
are fused directly to one another. In another embodiment, the PEST amino acid
sequence-
containing peptide and heterologous antigen are attached via a linker peptide.
In another
embodiment, the PEST amino acid sequence-containing peptide and heterologous
antigen are
attached via a heterologous peptide. In another embodiment, the PEST amino
acid sequence-
containing peptide is N-terminal to the heterologous antigen. In another
embodiment, the
PEST amino acid sequence-containing peptide is the N-terminal-most portion of
the fusion
protein.
[0065] In another embodiment, the present invention provides a method for
vaccinating a
human subject against an HPV, comprising the step of administering to the
subject the
recombinant Listeria strain provided herein, wherein the Listeria expresses an
HPV E7
antigen and wherein the Listeria expresses a mutant PrfA gene. In another
embodiment, the
mutant PrfA gene is a D133V PrfA mutation. In another embodiment, the mutant
PrfA gene is
in a plasmid in said recombinant Listeria. In another embodiment, the
recombinant Listeria
strain expresses the recombinant polypeptide. In another embodiment, the
recombinant
Listeria strain comprises a plasmid that encodes the recombinant polypeptide.
In one
embodiment, the recombinant Listeria strain comprises a bivalent episomal
expression vector,
the vector comprising a first, and a second nucleic acid molecule encoding a
heterologous
antigenic polypeptide or a functional fragment thereof, wherein the first and
the second
nucleic acid molecules each encode the heterologous antigenic polypeptide or
functional
fragment thereof in an open reading frame with an N-terminal or truncated or
detoxified
Listeriolysin 0 protein (LLO), or a truncated ActA protein, or a PEST amino
acid sequence.
[0066] In one embodiment, the heterologous antigens expressed by bivalent
expression
vector are HPV16 E7 and HPV18 E7.
[0067] In one embodiment, the recombinant Listeria strain comprises a
trivalent episomal
expression vector, the vector comprising a first, second, and third nucleic
acid molecule, each
encoding a heterologous antigenic polypeptide or a functional fragment
thereof, wherein the
first through third nucleic acid molecules each encode the heterologous
antigenic polypeptide
or functional fragment thereof in an open reading frame with an N-terminal or
truncated or
14

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
detoxified Listeriolysin 0 protein (LLO, a truncated ActA protein, or a PEST
amino acid
sequence.
[0068] In one embodiment, the bivalent, trivalent or quadravalent recombinant
Listeria
strains disclosed herein express at least one heterologous antigen from an
open reading frame
in a extrachromosomal plasmid or episome. In another embodiment, the bivalent,
or trivalent
recombinant Listeria strains disclosed herein express at least one
heterologous antigen from
an open reading frame ffrom at least one extrachromosomal plasmid or episome.
In another
embodiment, the bivalent recombinant Listeria strains disclosed herein express
two
heterologous antigens each from an open reading frame of two extrachromosomal
plasmids or
episomes. In another embodiment, the trivalent recombinant Listeria strains
disclosed herein
express three heterologous antigens each from an open reading frame of three
extrachromosomal plasmids or episomes. In another embodiment, the quadravalent

recombinant Listeria strains disclosed herein express four heterologous
antigens each from an
open reading frame of four extrachromosomal plasmids or episomes.
[0069] In another embodiment, the bivalent, trivalent, or quadravalent
recombinant Listeria
strains disclosed herein express at least one heterologous antigen from an
open reading frame
in the genome of the Listeria. In another embodiment, the bivalent, trivalent,
or quadravalent
recombinant Listeria strains provided herein express at least one heterologous
antigen from
both, an extrachromosomal plasmid or episome, and from the genome of a
Listeria provided
herein. In another embodiment, each heterologous antigen is expressed in a
fusion protein
with a PEST-containing polypeptide or peptide provided herein.
[0070] In one embodiment, bivalent and multivalent recombinant Listeria
encompassed by
the present invention include those described in US Pub. No. 2011/0129499, and
in US Pub
No. 2012/0135033, both of which are incorporated by reference in their
entirety herein.
[0071] In another embodiment, the subject is at risk for developing an HPV-
mediated
carcinogenesis (e.g. a cervical cancer). In another embodiment, the subject is
HPV-positive.
In another embodiment, the subject exhibits cervical intraepithelial
neoplasia. In another
embodiment, the subject exhibits a squamous intraepithelial lesion. In another
embodiment,
the subject exhibits a dysplasia in the cervix.
[0072] In one embodiment, there heterologous antigen is any tumor associated
antigen
known in the art and provided herein. In another embodiment, the heterologous
antigen is an
autoimmune antigen. In another embodiment, the heterologous antigen is an
infectious
disease antigen. In another embodiment, the heterologous antigen is an HPV-
related antigen.

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0073] The HPV that is the target of methods disclosed herein is, in another
embodiment, an
HPV 16. In another embodiment, the HPV is an HPV-18. In another embodiment,
the HPV is
selected from HPV-16 and HPV-18. In another embodiment, the HPV is an HPV-31.
In
another embodiment, the HPV is an HPV-35. In another embodiment, the HPV is an
HPV-39.
In another embodiment, the HPV is an HPV-45. In another embodiment, the HPV is
an HPV-
51. In another embodiment, the HPV is an HPV-52. In another embodiment, the
HPV is an
HPV-58. In another embodiment, the HPV is a high-risk HPV type. In another
embodiment,
the HPV is a mucosal HPV type.
[0074] In another embodiment, the present invention provides a method of
vaccinating a
human subject against an antigen of interest, the method comprising the step
of administering
intravenously to the human subject a recombinant Listeria strain comprising or
expressing the
antigen of interest, wherein the first peptide is selected from (a) an N-
terminal fragment of an
LLO protein; (b) an ActA protein or N-terminal fragment thereof; and (c) a
PEST amino acid
sequence-containing peptide, thereby vaccinating a human subject against an
antigen of
interest.
[0075] In another embodiment, the present invention provides a method of
vaccinating a
human subject against an antigen of interest, the method comprising the step
of administering
intravenously to the human subject an immunogenic composition, comprising a
fusion of a
first peptide to the antigen of interest, wherein the first peptide is
selected from (a) an N-
terminal fragment of an LLO protein; (b) an ActA protein or N-terminal
fragment thereof; and
(c) a PEST amino acid sequence-containing peptide, thereby vaccinating a human
subject
against an antigen of interest.
[0076] In another embodiment, the present invention provides a method of
vaccinating a
human subject against an antigen of interest, the method comprising the step
of administering
intravenously to the human subject a recombinant Listeria strain comprising a
recombinant
polypeptide, the recombinant polypeptide comprising a first peptide fused to
the antigen of
interest, wherein the first peptide is selected from (a) an N-terminal
fragment of an LLO
protein; (b) an ActA protein or N-terminal fragment thereof; and (c) a PEST
amino acid
sequence-containing peptide, thereby vaccinating a human subject against an
antigen of
interest.
[0077] In another embodiment, the present invention provides a method of
inducing a CTL
response in a human subject against an antigen of interest, the method
comprising the step of
administering to the human subject a recombinant Listeria strain comprising or
expressing the
16

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
antigen of interest, thereby inducing a CTL response in a human subject
against an antigen of
interest. In another embodiment, the step of administering is intravenous
administration.
[0078] As disclosed herein, recombinant Listeria strains expressing LLO-
antigen fusions
induce anti-tumor immunity (Example 1), elicit antigen-specific T cell
proliferation (Example
2), generate antigen-specific, and tumor-infiltrating T cells (Example 3).
Thus, vaccines
disclosed herein are efficacious at inducing immune responses against E7 and
E6.
[0079] In another embodiment, the present invention provides a method for
inducing a
regression of a cancer in a subject, comprising the step of administering to
the subject the
recombinant Listeria strain provided herein
[0080] In another embodiment, the present invention provides a method for
reducing an
incidence of relapse of a cancer in a subject, comprising the step of
administering to the
subject the recombinant Listeria strain provided herein.
[0081] In another embodiment, the present invention provides a method for
suppressing a
formation of a tumor in a subject, comprising the step of administering to the
subject the
recombinant Listeria strain provided herein.
[0082] In another embodiment, the present invention provides a method for
inducing a
remission of a cancer in a subject, comprising the step of administering to
the subject the
recombinant Listeria strain provided herein.
[0083] In another embodiment, the present invention provides a method for
impeding a
growth of a tumor in a human subject, comprising the step of administering to
the subject the
recombinant Listeria strain provided herein.
[0084] In another embodiment, the present invention provides a method for
reducing a size of
a tumor in a subject, comprising the step of administering to the subject the
recombinant
Listeria strain provided herein.
[0085] In one embodiment, the disease is an infectious disease, an autoimmune
disease, a
respiratory disease, a pre-cancerous condition or a cancer.
[0086] It will be well appreciated by the skilled artisan that the term "pre-
cancerous
condition" may encompass dysplasias, preneoplastic nodules; macroregenerative
nodules
(MRN); low-grade dysplastic nodules (LG-DN); high-grade dysplastic nodules (HG-
DN);
biliary epithelial dysplasia; foci of altered hepatocytes (FAH); nodules of
altered hepatocytes
(NAH); chromosomal imbalances; aberrant activation of telomerase; re-
expression of the
catalytic subunit of telomerase; expression of endothelial cell markers such
as CD31, CD34,
and BNH9 (see, e.g., Terracciano and Tomillo (2003) Pathologica 95:71-82; Su
and Bannasch
(2003) Toxicol. Pathol. 31:126-133; Rocken and Carl-McGrath (2001) Dig. Dis.
19:269-278;
17

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
Kotoula, et al. (2002) Liver 22:57-69; Frachon, et al. (2001) J. Hepatol.
34:850-857;
Shimonishi, et al. (2000) J. Hepatobiliary Pancreat. Surg. 7:542-550;
Nakanuma, et al. (2003)
J. Hepatobiliary Pancreat. Surg. 10:265-281). Methods for diagnosing cancer
and dysplasia
are disclosed (see, e.g., Riegler (1996) Semin. Gastrointest. Dis. 7:74-87;
Benvegnu, et al.
(1992) Liver 12:80-83; Giannini, et al. (1987) Hepatogastroenterol. 34:95-97;
Anthony (1976)
Cancer Res. 36:2579-2583).
[0087] In one embodiment, an infectious disease is one caused by, but not
limited to, any one
of the following pathogens: BCG/Tuberculosis, Malaria, Plasmodium falciparum,
plasmodium malariae, plasmodium vivax, Rotavirus, Cholera, Diptheria-Tetanus,
Pertussis,
Haemophilus influenzae, Hepatitis B, Human papilloma virus, Influenza
seasonal), Influenza
A (H1N1) Pandemic, Measles and Rubella, Mumps, Meningococcus A+C, Oral Polio
Vaccines, mono, bi and trivalent, Pneumococcal, Rabies, Tetanus Toxoid, Yellow
Fever,
Bacillus anthracis (anthrax), Clostridium botulinum toxin (botulism), Yersinia
pestis (plague),
Variola major (smallpox) and other related pox viruses, Francisella tularensis
(tularemia),
Viral hemorrhagic fevers, Arenaviruses (LCM, Junin virus, Machupo virus,
Guanarito virus,
Lassa Fever), Bunyaviruses (Hantaviruses, Rift Valley Fever), Flaviruses
(Dengue),
Filoviruses (Ebola , Marburg), Burkholderia pseudomallei, Coxiella burnetii (Q
fever),
Brucella species (brucellosis), Burkholderia mallei (glanders), Chlamydia
psittaci
(Psittacosis), Ricin toxin (from Ricinus communis), Epsilon toxin of
Clostridium perfringens,
Staphylococcus enterotoxin B, Typhus fever (Rickettsia prowazekii), other
Rickettsias, Food-
and Waterborne Pathogens, Bacteria (Diarrheagenic E.coli, Pathogenic Vibrios,
Shigella
species, Salmonella BCG/, Campylobacter jejuni, Yersinia enterocolitica),
Viruses
(Caliciviruses, Hepatitis A, West Nile Virus, LaCrosse, California
encephalitis, VEE, EEE,
WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Nipah virus,
hantaviruses,
Tickborne hemorrhagic fever viruses, Chikungunya virus, Crimean-Congo
Hemorrhagic fever
virus, Tickbome encephalitis viruses, Hepatitis B virus, Hepatitis C virus,
Herpes Simplex
virus (HSV), Human immunodeficiency virus (HIV), Human papillomavirus (HPV)),
Protozoa (Cryptosporidium parvum, Cyclospora cayatanensis, Giardia lamblia,
Entamoeba
histolytica, Toxoplasma), Fungi (Microsporidia), Yellow fever, Tuberculosis,
including drug-
resistant TB, Rabies, Prions, Severe acute respiratory syndrome associated
coronavirus
(SARS-CoV), Coccidioides posadasii, Coccidioides immitis, Bacterial vaginosis,
Chlamydia
trachomatis, Cytomegalovirus, Granuloma inguinale, Hemophilus ducreyi,
Neisseria
gonorrhea, Treponema pallidum, Trichomonas vaginalis, or any other infectious
disease
known in the art that is not listed herein.
18

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[0088] In another embodiment, the infectious disease is a livestock infectious
disease. In
another embodiment, livestock diseases can be transmitted to man and are
called "zoonotic
diseases." In another embodiment, these diseases include, but are not limited
to, Foot and
mouth disease, West Nile Virus, rabies, canine parvovirus, feline leukemia
virus, equine
influenza virus, infectious bovine rhinotracheitis (IBR), pseudorabies,
classical swine fever
(CSF), IBR, caused by bovine herpesvirus type 1 (BHV-1) infection of cattle,
and
pseudorabies (Aujeszky's disease) in pigs, toxoplasmosis, anthrax, vesicular
stomatitis virus,
rhodococcus equi, Tularemia, Plague (Yersinia pestis), trichomonas.
[0089] In another embodiment, the disease provided herein is a respiratory or
inflammatory
disease. In another embodiment, the respiratory or inflammatory disease is
chronic
obstructive pulmonary disease (COPD). In another embodiment, the disease is
asthma.
[0090] In one embodiment, live attenuated Listeria strains are capable of
alleviating asthma
symptoms without co-administration of other therapeutic agents, such as anti-
inflammatory
agents or bronchodilators. In another embodiment, the methods provided herein
further
comprise the step of co-administering to a subject the live attenuated
Listeria strain and one
or more therapeutic agents. In another embodiment, the therapeutic agent is an
anti-asthmatic
agent. In another embodiment, the agent is an anti-inflammatory agent, a non-
steroidal anti-
inflammatory agent, an antibiotic, an antichlolinerginc agent, a
bronchodilator, a
corticosteroid, a short-acting beta-agonist, a long-acting beta-agonist,
combination inhalers,
an antihistamine, or combinations thereof.
[0091] In one embodiment, the disease provided herein is a cancer or a tumor.
In one
embodiment, the tumor is cancerous. In another embodiment, the cancer is
breast cancer. In
another embodiment, the cancer is a cervical cancer. In another embodiment,
the cancer is a
Her2 containing cancer. In another embodiment, the cancer is a melanoma. In
another
embodiment, the cancer is pancreatic cancer. In another embodiment, the cancer
is ovarian
cancer. In another embodiment, the cancer is gastric cancer. In another
embodiment, the
cancer is a carcinomatous lesion of the pancreas. In another embodiment, the
cancer is
pulmonary adenocarcinoma. In another embodiment, it is a glioblastoma
multiforme. In
another embodiment, the cancer is colorectal adenocarcinoma. In another
embodiment, the
cancer is pulmonary squamous adenocarcinoma. In another embodiment, the cancer
is gastric
adenocarcinoma. In another embodiment, the cancer is an ovarian surface
epithelial neoplasm
(e.g. a benign, proliferative or malignant variety thereof). In another
embodiment, the cancer
is an oral squamous cell carcinoma. In another embodiment, the cancer is non-
small-cell lung
carcinoma. In another embodiment, the cancer is an endometrial carcinoma. In
another
19

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
embodiment, the cancer is a bladder cancer. In another embodiment, the cancer
is a head and
neck cancer. In another embodiment, the cancer is a prostate carcinoma. In
another
embodiment, the cancer is oropharyngeal cancer. In another embodiment, the
cancer is lung
cancer. In another embodiment, the cancer is anal cancer. In another
embodiment, the cancer
is colorectal cancerIn another embodiment, the cancer is vaginal cancer. In
another
embodiment, the cancer is esophageal cancer. The cervical tumor targeted by
methods
disclosed herein is, in another embodiment, a squamous cell carcinoma. In
another
embodiment, the cervical tumor is an adenocarcinoma. In another embodiment,
the cervical
tumor is an adenosquamous carcinoma. In another embodiment, the cervical tumor
is a small
cell carcinoma. In another embodiment, the cervical tumor is any other type of
cervical tumor
known in the art.
[0092] In one embodiment, the disease provided herein is a neoplasia. In
another
embodiment, the neoplasia is anal intraepithelial neoplasia (AIN). In another
embodiment, the
neoplasia is vaginal intraepithelial neoplasia (VIN).
[0093] The cervical tumor targeted by methods disclosed herein is, in another
embodiment, a
squamous cell carcinoma. In another embodiment, the cervical tumor is an
adenocarcinoma.
In another embodiment, the cervical tumor is an adenosquamous carcinoma. In
another
embodiment, the cervical tumor is a small cell carcinoma. In another
embodiment, the
cervical tumor is any other type of cervical tumor known in the art.
[0094] In one embodiment, the antigen provided herein is a heterologous tumor
antigen,
which is also referred to herein as "tumor antigen" "antigenic polypeptide,"
or "foreign
antigen." In another embodiment, the antigen is Human Papilloma Virus-E7 (HPV-
E7)
antigen, which in one embodiment, is from HPV16 (in one embodiment, GenBank
Accession
No. AAD33253) and in another embodiment, from HPV18 (in one embodiment,
GenBank
Accession No. P06788). In another embodiment, the antigenic polypeptide is HPV-
E6, which
in one embodiment, is from HPV16 (in one embodiment, GenBank Accession No.
AAD33252, AAM51854, AAM51853, or AAB67615) and in another embodiment, from
HPV18 (in one embodiment, GenBank Accession No. P06463). In another
embodiment, the
antigenic polypeptide is a Her/2-neu antigen. In another embodiment, the
antigenic
polypeptide is Prostate Specific Antigen (PSA) (in one embodiment, GenBank
Accession No.
CAD30844, CAD54617, AAA58802, or NP_001639). In another embodiment, the
antigenic
polypeptide is Stratum Corneum Chymotryptic Enzyme (SCCE) antigen (in one
embodiment,
GenBank Accession No. AAK69652, AAK69624, AAG33360, AAF01139, or AAC37551).
In another embodiment, the antigenic polypeptide is Wilms tumor antigen 1,
which in another

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
embodiment is WT-1 Telomerase (GenBank Accession. No. P49952, P22561,
NP_659032,
CAC39220.2, or EAW68222.1). In another embodiment, the antigenic polypeptide
is hTERT
or Telomerase (GenBank Accession. No. NM003219 (variant 1), NM198255 (variant
2), NM
198253 (variant 3), or NM 198254 (variant 4). In another embodiment, the
antigenic
polypeptide is Proteinase 3 (in one embodiment, GenBank Accession No. M29142,
M75154,
M96839, X55668, NM 00277, M96628 or X56606). In another embodiment, the
antigenic
polypeptide is Tyrosinase Related Protein 2 (TRP2) (in one embodiment, GenBank
Accession
No. NP_001913, ABI73976, AAP33051, or Q95119). In another embodiment, the
antigenic
polypeptide is High Molecular Weight Melanoma Associated Antigen (HMW-MAA) (in
one
embodiment, GenBank Accession No. NP_001888, AAI28111, or AAQ62842). In
another
embodiment, the antigenic polypeptide is Testisin (in one embodiment, GenBank
Accession
No. AAF79020, AAF79019, AAG02255, AAK29360, AAD41588, or NP_659206). In
another embodiment, the antigenic polypeptide is NY-ES 0-1 antigen (in one
embodiment,
GenBank Accession No. CAA05908, P78358, AAB49693, or NP_640343). In another
embodiment, the antigenic polypeptide is PSCA (in one embodiment, GenBank
Accession
No. AAH65183, NP_005663, NP_082492, 043653, or CAB97347). In another
embodiment,
the antigenic polypeptide is Interleukin (IL) 13 Receptor alpha (in one
embodiment, GenBank
Accession No. NP_000631, NP_001551, NP_032382, NP_598751, NP_001003075, or
NP_999506). In another embodiment, the antigenic polypeptide is Carbonic
anhydrase IX
(CAIX) (in one embodiment, GenBank Accession No. CAI13455, CAI10985, EAW58359,
NP_001207, NP_647466, or NP_001101426). In another embodiment, the antigenic
polypeptide is carcinoembryonic antigen (CEA) (in one embodiment, GenBank
Accession
No. AAA66186, CAA79884, CAA66955, AAA51966, AAD15250, or AAA51970.). In
another embodiment, the antigenic polypeptide is MAGE-A (in one embodiment,
GenBank
Accession No. NP_786885, NP_786884, NP_005352, NP_004979, NP_005358, or NP_
005353). In another embodiment, the antigenic polypeptide is survivin (in one
embodiment,
GenBank Accession No. AAC51660, AAY15202, ABF60110, NP_001003019, or NP_
001082350). In another embodiment, the antigenic polypeptide is GP100 (in one
embodiment,
GenBank Accession No. AAC60634, YP_655861, or AAB31176). In another
embodiment,
the antigenic polypeptide is any other antigenic polypeptide known in the art.
In another
embodiment, the antigenic peptide of the compositions and methods disclosed
herein
comprise an immunogenic portion of the antigenic polypeptide.
1100951 In another embodiment, the antigen is HPV-E6. In another embodiment,
the antigen is
telomerase (TERT). In another embodiment, the antigen is LMP-1. In another
embodiment,
21

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
the antigen is p53. In another embodiment, the antigen is mesothelin. In
another embodiment,
the antigen is EGFRVIII. In another embodiment, the antigen is carboxic
anhydrase IX
(CAIX). In another embodiment, the antigen is PSMA. In another embodiment, the
antigen is
HMW-MAA. In another embodiment, the antigen is HIV-1 Gag. In another
embodiment, the
antigen is Tyrosinase related protein 2. In another embodiment, the antigen is
selected from
HPV-E7, HPV-E6, Her-2, HIV-1 Gag, LMP-1, p53, PSMA, carcinoembryonic antigen
(CEA), LMP-1,kallikrein-related peptidase 3 (KLK3), KLK9, Muc, Tyrosinase
related protein
2, Mud, FAP, IL-13R alpha 2, PSA (prostate-specific antigen), gp-100, heat-
shock protein
70 (HSP-70), beta-HCG, EGFR-III, Granulocyte colony-stimulating factor (G-
CSF),
Angiogenin, Angiopoietin-1, Del-1, Fibroblast growth factors: acidic (aFGF) or
basic (bFGF),
Follistatin, Granulocyte colony-stimulating factor (G-CSF), Hepatocyte growth
factor
(HGF)/scatter factor (SF), Interleukin-8 (IL-8), Leptin, Midkine, Placental
growth factor,
Platelet-derived endothelial cell growth factor (PD-ECGF), Platelet-derived
growth factor-BB
(PDGF-BB), Pleiotrophin (PTN), Progranulin, Proliferin, Transforming growth
factor-alpha
(TGF-alpha), Transforming growth factor-beta (TGF-beta), Tumor necrosis factor-
alpha
(TNF-alpha), Vascular endothelial growth factor (VEGF)/vascular permeability
factor (VPF),
VEGFR, VEGFR2 (KDR/FLK-1) or a fragment thereof, FLK-1 or an epitope thereof,
FLK-
El , FLK-E2, FLK-Ii, endoglin or a fragment thereof, Neuropilin 1 (NRP-1),
Angiopoietin 1
(Angl), Tie2, Platelet-derived growth factor (PDGF), Platelet-derived growth
factor receptor
(PDGFR), Transforming growth factor-beta (TGF-13), endoglin, TGF-r= receptors,
monocyte
chemotactic protein-1 (MCP-1), VE-cadherin, CD31, ephrin, ICAM-1, V-CAM-1, VAP-
1, E-
selectin, plasminogen activators, plasminogen activator inhibitor-1, Nitric
oxide synthase
(NOS), COX-2, AC133, or Idl/Id3, Angiopoietin 3, Angiopoietin 4, Angiopoietin
6, CD105,
EDG, HHT1, ORW, ORW1 or a TGFbeta co-receptor, or a combination thereof. In
another
embodiment, the antigen is a chimeric Her2/neu antigen as disclosed in US
Patent Application
Publication No. 2011/0142791, which is incorporated by reference herein in its
entirety. The
use of fragments of antigens provided herein is also encompassed by the
present invention.
[0096] In another embodiment, the heterologous tumor antigen provided herein
is a tumor-
associated antigen, which in one embodiment, is one of the following tumor
antigens: a
MAGE (Melanoma-Associated Antigen E) protein, e.g. MAGE 1, MAGE 2, MAGE 3,
MAGE 4, a tyrosinase; a mutant ras protein; a mutant p53 protein; p97 melanoma
antigen, a
ras peptide or p53 peptide associated with advanced cancers; the HPV 16/18
antigens
associated with cervical cancers, KLH antigen associated with breast
carcinoma, CEA
(carcinoembryonic antigen) associated with colorectal cancer, a MARTI antigen
associated
22

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
with melanoma, or the PSA antigen associated with prostate cancer. In another
embodiment,
the antigen for the compositions and methods provided herein are melanoma-
associated
antigens, which in one embodiment are TRP-2, MAGE-1, MAGE-3, gp-100,
tyrosinase, HSP-
70, beta-HCG, or a combination thereof. It is to be understood that a skilled
artisan would be
able to use any heterologous antigen not mentioned herein but known in the art
for use in the
methods and compositions provided herein. It is also to be understood that the
present
invention provides, but is not limited by, an attenuated Listeria comprising a
nucleic acid that encodes
at least one of the antigens disclosed herein. The present invention
encompasses nucleic acids
encoding mutants, muteins, splice variants, fragments, truncated variants,
soluble variants,
extracellular domains, intracellular domains, mature sequences, and the like,
of the disclosed antigens.
Provided are nucleic acids encoding epitopes, oligo- and polypeptides of these
antigens. Also provided
are codon optimized embodiments, that is, optimized for expression in
Listeria. The cited references,
GenBank Acc. Nos., and the nucleic acids, peptides, and polypeptides disclosed
herein, are all
incorporated herein by reference in their entirety. In another embodiment, the
selected nucleic
acid sequence can encode a full length or a truncated gene, a fusion or tagged
gene, and can
be a cDNA, a genomic DNA, or a DNA fragment, preferably, a cDNA. It can be
mutated or
otherwise modified as desired. These modifications include codon optimizations
to optimize
codon usage in the selected host cell or bacteria, i.e. Listeria. The selected
sequence can also
encode a secreted, cytoplasmic, nuclear, membrane bound or cell surface
polypeptide.
[0097] In one embodiment, vascular endothelial growth factor (VEGF) is an
important
signaling protein involved in both vasculogenesis (the formation of the
embryonic circulatory
system) and angiogenesis (the growth of blood vessels from pre-existing
vasculature). In one
embodiment, VEGF activity is restricted mainly to cells of the vascular
endothelium, although
it does have effects on a limited number of other cell types (e.g. stimulation
monocyte/macrophage migration). In vitro, VEGF has been shown to stimulate
endothelial
cell mitogenesis and cell migration. VEGF also enhances microvascular
permeability and is
sometimes referred to as vascular permeability factor.
[0098] In one embodiment, all of the members of the VEGF family stimulate
cellular responses
by binding to tyrosine kinase receptors (the VEGFRs) on the cell surface,
causing them to
dimerize and become activated through transphosphorylation. The VEGF receptors
have an
extracellular portion consisting of 7 immunoglobulin-like domains, a single
transmembrane
spanning region and an intracellular portion containing a split tyrosine-
kinase domain.
[0099] In one embodiment, VEGF-A is a VEGFR-2 (KDR/Flk-1) ligand as well as a
VEGFR-1
(Flt-1) ligand. In one embodiment, VEGFR- mediates almost all of the known
cellular responses
23

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
to VEGF. The function of VEGFR-1 is less well defined, although it is thought
to modulate
VEGFR-2 signaling, in one embodiment, via sequestration of VEGF from VEGFR-2
binding,
which in one embodiment, is particularly important during vasculogenesis in
the embryo. In one
embodiment, VEGF-C and VEGF-D are ligands of the VEGFR-3 receptor, which in
one
embodiment, mediates lymphangiogenesis.
[00100] In one embodiment, the compositions disclosed herein comprise a VEGF
receptor or a
fragment thereof, which in one embodiment, is a VEGER-2 and, in another
embodiment, a
VEGFR-1, and, in another embodiment, VEGER-3.
[00101] In one embodiment, vascular Endothelial Growth Factor Receptor 2
(VEGER2) is
highly expressed on activated endothelial cells (ECs) and participates in the
formation of new
blood vessels. In one embodiment, VEGER2 binds all 5 isoforms of VEGF. In one
embodiment, signaling of VEGF through VEGER2 on ECs induces proliferation,
migration, and
eventual differentiation. In one embodiment, the mouse homologue of VEGER2 is
the fetal liver
kinase gene-1 (Hk-1), which is a strong therapeutic target, and has important
roles in tumor
growth, invasion, and metastasis. In one embodiment, VEGFR2 is also referred
to as kinase
insert domain receptor (a type III receptor tyrosine kinase) (KDR), cluster of
differentiation 309
(CD309), FLK1, Ly73, Krd-1, VEGER, VEGER-2, or 6130401C07.
[00102] In other embodiments, the antigen is derived from a fungal pathogen,
bacteria, parasite,
helminth, or viruses. In other embodiments, the antigen is selected from
tetanus toxoid,
hemagglutinin molecules from influenza virus, diphtheria toxoid, HIV gp120,
HIV gag protein,
IgA protease, insulin peptide B, Spongospora subterranea antigen, vibriose
antigens, Salmonella
antigens, pneumococcus antigens, respiratory syncytial virus antigens,
Haemophilus influenza
outer membrane proteins, Helicobacter pylori urease, Neisseria meningitidis
pilins, N.
gonorrhoeae pilins, the melanoma-associated antigens (TRP-2, MAGE-1, MAGE-3,
gp-100,
tyrosinase, MART-1, HSP-70, beta-HCG), human papilloma virus antigens El and
E2 from type
HPV-16, -18, -31, -33, -35 or -45 human papilloma viruses, the tumor antigens
CEA, the ras
protein, mutated or otherwise, the p53 protein, mutated or otherwise, Mud, or
pSA.
[00103]In other embodiments, the antigen is associated with one of the
following diseases;
cholera, diphtheria, Haemophilus, hepatitis A, hepatitis B, influenza,
measles, meningitis,
mumps, pertussis, small pox, pneumococcal pneumonia, polio, rabies, rubella,
tetanus,
tuberculosis, typhoid, Varicella-zoster, whooping cough3 yellow fever, the
immunogens and
antigens from Addison's disease, allergies, anaphylaxis, Bruton's syndrome,
cancer, including
solid and blood borne tumors, eczema, Hashimoto's thyroiditis, polymyositis,
dermatomyositis, type 1 diabetes mellitus, acquired immune deficiency
syndrome, transplant
24

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
rejection, such as kidney, heart, pancreas, lung, bone, and liver transplants,
Graves disease,
polyendocrine autoimmune disease, hepatitis, microscopic polyarteritis,
polyarteritis nodosa,
pemphigus, primary biliary cirrhosis, pernicious anemia, coeliac disease,
antibody-mediated
nephritis, glomerulonephritis, rheumatic diseases, systemic lupus
erthematosus, rheumatoid
arthritis, seronegative spondylarthritides, rhinitis, sjogren's syndrome,
systemic sclerosis,
sclerosing cholangitis, Wegener's granulomatosis, dermatitis herpetiformis,
psoriasis, vitiligo,
multiple sclerosis, encephalomyelitis, Guillain-Barre syndrome, myasthenia
gravis, Lambert-
Eaton syndrome, sclera, episclera, uveitis, chronic mucocutaneous candidiasis,
urticaria,
transient hypogammaglobulinemia of infancy, myeloma, X-linked hyper IgM
syndrome,
Wiskott-Aldrich syndrome, ataxia telangiectasia, autoimmune hemolytic anemia,
autoimmune
thrombocytopenia, autoimmune neutropenia, Waldenstrom's macroglobulinemia,
amyloidosis, chronic lymphocytic leukemia, non-Hodgkin's lymphoma, malarial
circumsporozite protein, microbial antigens, viral antigens, autoantigens, and
lesteriosis.
1001041In another embodiment, an HPV E6 antigen is utilized instead of or in
addition to an
E7 antigen in a method disclosed herein for treating, protecting against, or
inducing an
immune response against a cervical cancer.
1001051In another embodiment, an ActA protein fragment is utilized instead of
or in addition
to an LLO fragment in a method disclosed herein for treating, protecting
against, or inducing
an immune response against a cervical cancer.
1001061In another embodiment, a PEST amino acid sequence-containing protein
fragment is
utilized instead of or in addition to an LLO fragment in a method disclosed
herein for treating,
protecting against, or inducing an immune response against a cervical cancer.
1001071In another embodiment, disclosed herein is an immunogenic composition
comprising
a recombinant Listeria of the present invention. In another embodiment, the
immunogenic
composition of methods and compositions disclosed herein comprises a
recombinant vaccine
vector of the present invention. In another embodiment, the immunogenic
composition
comprises a plasmid of the present invention. In another embodiment, the
immunogenic
composition comprises an adjuvant. In one embodiment, a vector disclosed
herein may be
administered as part of a vaccine composition.]
1001081In another embodiment, a vaccine disclosed herein is delivered with an
adjuvant. In
one embodiment, the adjuvant favors a predominantly Thl -mediated immune
response. In
another embodiment, the adjuvant favors a ml-type immune response. In another
embodiment, the adjuvant favors a Thl -mediated immune response. In another
embodiment,
the adjuvant favors a cell-mediated immune response over an antibody-mediated
response. In

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
another embodiment, the adjuvant is any other type of adjuvant known in the
art. In another
embodiment, the immunogenic composition induces the formation of a T cell
immune
response against a target protein present on a tumor cell.
11001091 In another embodiment, the present invention provides a method for
inducing an anti-
E7 cytotoxic T cell (CTL) response in a human subject, comprising the step of
administering
to the subject a recombinant Listeria strain, the recombinant Listeria strain
comprising a
recombinant polypeptide comprising an N-terminal fragment of an LLO protein
and an HPV
E7 antigen, thereby inducing an anti-E7 CTL response in a human subject. In
another
embodiment, the recombinant Listeria strain comprises a plasmid that encodes
the
recombinant polypeptide. In another embodiment, the method further comprises
the step of
boosting the subject with a recombinant Listeria strain of the present
invention. In another
embodiment, the method further comprises the step of boosting the subject with
an
immunogenic composition comprising an E7 antigen. In another embodiment, the
method
further comprises the step of boosting the subject with an immunogenic
composition that
directs a cell of the subject to express an E7 antigen. In another embodiment,
the CTL
response is capable of therapeutic efficacy against an HPV-mediated disease,
disorder, or
symptom. In another embodiment, the CTL response is capable of prophylactic
efficacy
against an HPV-mediated disease, disorder, or symptom.
11001101 In another embodiment, the present invention provides a method of
treating or
ameliorating an HPV-mediated disease, disorder, or symptom in a subject,
comprising the
step of administering to the subject a recombinant Listeria strain, the
recombinant Listeria
strain comprising a recombinant polypeptide comprising an N-terminal fragment
of an LLO
protein and an HPV E7 antigen, whereby the recombinant Listeria strain induces
an immune
response against the E7 antigen, thereby treating or ameliorating an HPV-
mediated disease,
disorder, or symptom in a subject. In another embodiment, the subject is a
human subject. In
another embodiment, the subject is a non-human mammal. In another embodiment,
the
subject is any other type of subject known in the art.
[00111]The HPV causing the disease, disorder, or symptom is, in another
embodiment, an
HPV 16. In another embodiment, the HPV is an HPV-18. In another embodiment,
the HPV is
an HPV-31. In another embodiment, the HPV is an HPV-35. In another embodiment,
the
HPV is an HPV-39. In another embodiment, the HPV is an HPV-45. In another
embodiment,
the HPV is an HPV-51. In another embodiment, the HPV is an HPV-52. In another
embodiment, the HPV is an HPV-58. In another embodiment, the HPV is a high-
risk HPV
type. In another embodiment, the HPV is a mucosal HPV type.
26

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
11001121 In another embodiment, the HPV-mediated disease, disorder, or symptom
is genital
warts. In another embodiment, the HPV-mediated disease, disorder, or symptom
is non-
genital warts. In another embodiment, the HPV-mediated disease, disorder, or
symptom is a
respiratory papilloma. In another embodiment, the HPV-mediated disease,
disorder, or
symptom is any other HPV-mediated disease, disorder, or symptom known in the
art.
11001131 In another embodiment, an HPV E6 antigen is utilized instead of or in
addition to an
E7 antigen in a method disclosed herein for treating or ameliorating an HPV-
mediated
disease, disorder, or symptom.
11001141 In another embodiment, an ActA protein fragment is utilized instead
of or in addition
to an LLO fragment in a method disclosed herein for treating or ameliorating
an HPV-
mediated disease, disorder, or symptom.
11001151 In another embodiment, a PEST amino acid sequence-containing protein
fragment is
utilized instead of or in addition to an LLO fragment in a method disclosed
herein for treating
or ameliorating an HPV-mediated disease, disorder, or symptom.
11001161 In another embodiment, an HPV E6 antigen is utilized instead of or in
addition to an
E7 antigen in a method disclosed herein for treating or ameliorating an HPV-
mediated
disease, disorder, or symptom.
[00117] The antigen of methods and compositions disclosed herein is, in
another embodiment,
an HPV E7 protein. In another embodiment, the antigen is an HPV E6 protein. In
another
embodiment, the antigen is any other HPV protein known in the art.
[00118]"E7 antigen" refers, in another embodiment, to an E7 protein. In
another embodiment,
the term refers to an E7 fragment. In another embodiment, the term refers to
an E7 peptide. In
another embodiment, the term refers to any other type of E7 antigen known in
the art.
[00119] The E7 protein of methods and compositions disclosed herein is, in
another
embodiment, an HPV 16 E7 protein. In another embodiment, the E7 protein is an
HPV-18 E7
protein. In another embodiment, the E7 protein is an HPV-31 E7 protein. In
another
embodiment, the E7 protein is an HPV-35 E7 protein. In another embodiment, the
E7 protein
is an HPV-39 E7 protein. In another embodiment, the E7 protein is an HPV-45 E7
protein. In
another embodiment, the E7 protein is an HPV-51 E7 protein. In another
embodiment, the E7
protein is an HPV-52 E7 protein. In another embodiment, the E7 protein is an
HPV-58 E7
protein. In another embodiment, the E7 protein is an E7 protein of a high-risk
HPV type. In
another embodiment, the E7 protein is an E7 protein of a mucosal HPV type.
27

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
1001201"E6 antigen" refers, in another embodiment, to an E6 protein. In
another embodiment,
the term refers to an E6 fragment. In another embodiment, the term refers to
an E6 peptide. In
another embodiment, the term refers to any other type of E6 antigen known in
the art.
1001211The E6 protein of methods and compositions disclosed herein is, in
another
embodiment, an HPV 16 E6 protein. In another embodiment, the E6 protein is an
HPV-18 E6
protein. In another embodiment, the E6 protein is an HPV-31 E6 protein. In
another
embodiment, the E6 protein is an HPV-35 E6 protein. In another embodiment, the
E6 protein
is an HPV-39 E6 protein. In another embodiment, the E6 protein is an HPV-45 E6
protein. In
another embodiment, the E6 protein is an HPV-51 E6 protein. In another
embodiment, the E6
protein is an HPV-52 E6 protein. In another embodiment, the E6 protein is an
HPV-58 E6
protein. In another embodiment, the E6 protein is an E6 protein of a high-risk
HPV type. In
another embodiment, the E6 protein is an E6 protein of a mucosal HPV type.
1001221The immune response induced by methods and compositions disclosed
herein is, in
another embodiment, a T cell response. In another embodiment, the immune
response
comprises a T cell response. In another embodiment, the response is a CD8+ T
cell response.
In another embodiment, the response comprises a CD8+ T cell response.
1001231In one embodiment, compositions disclosed herein induce a strong innate
stimulation
of interferon-gamma, which in one embodiment, has anti-angiogenic properties.
In one
embodiment, a Listeria disclosed herein induces a strong innate stimulation of
interferon-
gamma, which in one embodiment, has anti-angiogenic properties (Dominiecki et
al., Cancer
Immunol Immunother. 2005 May;54(5):477-88. Epub 2004 Oct 6, incorporated
herein by
reference in its entirety; Beatty and Paterson, J Immunol. 2001 Feb
15;166(4):2276-82,
incorporated herein by reference in its entirety). In another embodiment,
methods disclosed
herein increase a level of interferon-gamma producing cells. In one
embodiment, anti-
angiogenic properties of Listeria are mediated by CD4+ T cells (Beatty and
Paterson, 2001).
In another embodiment, anti-angiogenic properties of Listeria are mediated by
CD8+ T cells.
In another embodiment, IFN-gamma secretion as a result of Listeria vaccination
is mediated
by NK cells, NKT cells, Thl CD4+ T cells, TC1 CD8+ T cells, or a combination
thereof.
1001241In another embodiment, compositions disclosed herein induce production
of one or
more anti-angiogenic proteins or factors. In one embodiment, the anti-
angiogenic protein is
IFN-gamma. In another embodiment, the anti-angiogenic protein is pigment
epithelium-
derived factor (PEDF); angiostatin; endostatin; fms-like tyrosine kinase
(sFlt)-1; or soluble
endoglin (sEng). In one embodiment, a Listeria disclosed herein is involved in
the release of
28

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
anti-angiogenic factors, and, therefore, in one embodiment, has a therapeutic
role in addition
to its role as a vector for introducing an antigen to a subject.
1001251In another embodiment, the administration of compositions disclosed
herein induces
robust systemic antigen-specific immunity. In another embodiment, the
administration of
compositions disclosed herein induces epitope spreading. In another
embodiment, the
administration of compositions disclosed herein induces broad-based response
to self-derived
tumor or neoplasia antigens. In another embodiment the immune response induced
by
methods and compositions disclosed herein comprises an improvement of the
overall balance
of suppressor and effector immune cells in the tumor or neoplasia
microenvironment (TME).
In another embodiment the immune response induced by methods and compositions
disclosed
herein comprises improvement in the systemic balance of suppressor and
effector
immunocytes.
1001261In one embodiment, compositions and methods of use thereof disclosed
herein
generate effector T cells that are able to infiltrate the tumor or neoplasia,
destroy tumor cells
and eradicate the disease. In another embodiment, methods of use of this
invention increase
tumor infiltration by T effector cells. In another embodiment, T effector
cells comprise CD8+
T cells. In another embodiment, T effector cells comprise CD4+ T cells.
1001271In one embodiment, tumor infiltrating lymphocytes (TILs) are associated
with better
prognosis in several tumors, such as colon, ovarian and melanoma. In colon
cancer, tumors
without signs of micrometastasis have an increased infiltration of immune
cells and a Thl
expression profile, which correlate with an improved survival of patients.
Moreover, the
infiltration of the tumor by T cells has been associated with success of
immunotherapeutic
approaches in both pre-clinical and human trials. In one embodiment, the
infiltration of
lymphocytes into the tumor site is dependent on the up-regulation of adhesion
molecules in
the endothelial cells of the tumor vasculature, generally by proinflammatory
cytokines, such
as IFN-7, TNF-a and IL-1. Several adhesion molecules have been implicated in
the process of
lymphocyte infiltration into tumors, including intercellular adhesion molecule
1 (ICAM-1),
vascular endothelial cell adhesion molecule 1 (V-CAM-1), vascular adhesion
protein 1 (VAP-
1) and E-selectin. However, these cell-adhesion molecules are commonly down-
regulated in
the tumor vasculature. Thus, in one embodiment, cancer vaccines As disclosed
herein
increase TILs, up-regulate adhesion molecules (in one embodiment, ICAM-1, V-
CAM-1,
VAP-1, E-selectin, or a combination thereof), up-regulate pro-inflammatory
cytokines (in one
embodiment, IFN-7, TNF-a, IL-1, or a combination thereof), or a combination
thereof.
29

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
1001281The N-terminal LLO protein fragment of methods and compositions
disclosed herein
comprises, in another embodiment, SEQ ID No: 2. In another embodiment, the
fragment
comprises an LLO signal peptide. In another embodiment, the fragment comprises
SEQ ID
No: 2. In another embodiment, the fragment consists approximately of SEQ ID
No: 2. In
another embodiment, the fragment consists essentially of SEQ ID No: 2. In
another
embodiment, the fragment corresponds to SEQ ID No: 2. In another embodiment,
the
fragment is homologous to SEQ ID No: 2. In another embodiment, the fragment is

homologous to a fragment of SEQ ID No: 2. The ALLO used in some of the
Examples was
416 AA long (exclusive of the signal sequence), as 88 residues from the amino
terminus
which is inclusive of the activation domain containing cysteine 484 were
truncated. It will be
clear to those skilled in the art that any ALLO without the activation domain,
and in particular
without cysteine 484, are suitable for methods and compositions of the present
invention. In
another embodiment, fusion of an E7 or E6 antigen to any ALLO, including the
PEST amino
acid AA sequence, SEQ ID NO: 1, enhances cell mediated and anti-tumor immunity
of the
antigen.
1001291The LLO protein utilized to construct vaccines disclosed herein has, in
another
embodiment, the sequence:
MKKIMLVFITLILVSLPIAQQTEAKDASAFNKENSISSMAPPASPPASPKTPIEK
KHADEIDKYIQGLDYNKNNVLVYHGDAVTNVPPRKGYKDGNEYIVVEKKKKSIN
QNNADIQVVNAIS S LTYPGALV KANS ELVEN QPDVLPVKRD S LTLSIDLPGMTNQ
DNKIVV KNATKS NVNNAVNTLVERWNEKYAQAYPNVS A KIDYDD EMAYS ES QL
IAKFGTAFKAVNNSLNVNFGAISEGKMQEEVISFKQIYYNVNVNEPTRPSRFFGKA
VTKEQLQALGVNAENPPAYIS S VAYGRQVYLKLS TNS HS TKVKAAFDAAVS GKS
VS GDVELTNIIKNS SFKAVIYGGS AKDEVQIID GNLGD LRD ILKKGATFNRETP GVP
IAYTTNFLKDNELAVIKNNSEYIETTS KAYTDGKINIDHSGGYVAQFNISWDEVNY
DPEGNEIVQHKNWSENNKSKLAHFTSSIYLPGNARNINVYAKECTGLAWEWWRT
VIDDRNLPLVKNRNISIWGTTLYPKYSNKVDNPIE (GenB ank Accession No.
P13128; SEQ ID NO: 3; nucleic acid sequence is set forth in GenBank Accession
No.
X15127). The first 25 AA of the proprotein corresponding to this sequence are
the signal
sequence and are cleaved from LLO when it is secreted by the bacterium. Thus,
in this
embodiment, the full length active LLO protein is 504 residues long. In
another
embodiment, the above LLO fragment is used as the source of the LLO fragment
incorporated in a vaccine of the present invention.

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
11001301 In another embodiment, the N-terminal fragment of an LLO protein
utilized in
compositions and methods disclosed herein has the sequence:
M KKIMLVFITLILVS LPIAQ QTEAKD ASAFNKENS IS S VAPPAS PPAS PKTPIEKK
HADEIDKYIQGLDYNKNNVLVYHGDAVTNVPPRKGYKD GNEYIVVEKKKKSINQ
NNADIQVVNAISSLTYPGALVKANSELVENQPDVLPVKRD S LTLS ID LPGMTNQD
NKIVVKNAT KS NVNNAVNTLVERWNEKYA QAYS NVS A KIDYDDEMAYS ES QLI
AKFGTAFKAVNNSLNVNFGAISEGKMQEEVISFKQIYYNVNVNEPTRPSRFFGKA
VT KEQLQALGVNAENPPAYIS S VAYGRQVYLKLS TNS HS TKVKAAFDAAVS GKS
VS GDVELTNIIKN S S FKAVIYGGS AKDEVQIID GNLGD LRD ILKKGATFNRETPGVP
IAYTTNFLKDNELAVIKNNSEYIETTS KAYTDGKINIDHSGGYVAQFNISWDEVNY
D (SEQ ID NO: 2).
11001311 In another embodiment, the LLO fragment corresponds to about AA 20-
442 of an
LLO protein utilized herein.
[00132] In another embodiment, the LLO fragment has the sequence:
M KKIMLVFITLILVS LPIAQ QTEAKD ASAFNKENS IS S VAPPAS PPAS PKTPIEKK
HADEIDKYIQGLDYNKNNVLVYHGDAVTNVPPRKGYKD GNEYIVVEKKKKSINQ
NNADIQVVNAISSLTYPGALVKANSELVENQPDVLPVKRD S LTLS ID LPGMTNQD
NKIVVKNAT KS NVNNAVNTLVERWNEKYA QAYS NVS A KIDYDDEMAYS ES QLI
AKFGTAFKAVNNSLNVNFGAISEGKMQEEVISFKQIYYNVNVNEPTRPSRFFGKA
VT KEQLQALGVNAENPPAYIS S VAYGRQVYLKLS TNS HS TKVKAAFDAAVS GKS
VS GDVELTNIIKN S S FKAVIYGGS AKDEVQIID GNLGD LRD ILKKGATFNRETPGV
PIAYTTNFLKDNELAVIKNNSEYIETTSKAYTD (SEQ ID NO: 4).
110013311n another embodiment, "truncated LLO" or "ALLO" refers to a fragment
of LLO that
comprises the PEST amino acid domain. In another embodiment, the terms refer
to an LLO
fragment that comprises a PEST sequence.
[00134] In another embodiment, the terms refer to an LLO fragment that does
not contain the
activation domain at the amino terminus and does not include cysteine 484. In
another
embodiment, the terms refer to an LLO fragment that is not hemolytic. In
another
embodiment, the LLO fragment is rendered non-hemolytic by deletion or mutation
of the
activation domain. In another embodiment, the LLO fragment is rendered non-
hemolytic by
deletion or mutation of cysteine 484. In another embodiment, the LLO fragment
is rendered
non-hemolytic by deletion or mutation at another location.
31

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[001351In another embodiment, the LLO fragment consists of about the first 441
AA of the
LLO protein. In another embodiment, the LLO fragment consists of about the
first 420 AA of
LLO. In another embodiment, the LLO fragment is a non-hemolytic form of the
LLO protein.
[001361In another embodiment, the LLO fragment contains residues of a
homologous LLO
protein that correspond to one of the above AA ranges. The residue numbers
need not, in
another embodiment, correspond exactly with the residue numbers enumerated
above; e.g. if
the homologous LLO protein has an insertion or deletion, relative to an LLO
protein utilized
herein, then the residue numbers can be adjusted accordingly.
11001371In another embodiment, the LLO fragment is any other LLO fragment
known in the
art.
[001381In another embodiment, the recombinant Listeria strain is administered
to the human
subject at a dose of 1 x 109 - 3.31 x 1010 CFU. In another embodiment, the
dose is 5-500 x 108
CFU. In another embodiment, the dose is 7-500 x 108 CFU. In another
embodiment, the dose
is 10-500 x 108 CFU. In another embodiment, the dose is 20-500 x 108 CFU. In
another
embodiment, the dose is 30-500 x 108 CFU. In another embodiment, the dose is
50-500 x 108
CFU. In another embodiment, the dose is 70-500 x 108 CFU. In another
embodiment, the dose
is 100-500 x 108 CFU. In another embodiment, the dose is 150-500 x 108 CFU. In
another
embodiment, the dose is 5-300 x 108 CFU. In another embodiment, the dose is 5-
200 x 108
CFU. In another embodiment, the dose is 5-150 x 108 CFU. In another
embodiment, the dose
is 5-100 x 108 CFU. In another embodiment, the dose is 5-70 x 108 CFU. In
another
embodiment, the dose is 5-50 x 108 CFU. In another embodiment, the dose is 5-
30 x 108 CFU.
In another embodiment, the dose is 5-20 x 108 CFU. In another embodiment, the
dose is 1-30
x 109 CFU. In another embodiment, the dose is 1-20 x 109 CFU. In another
embodiment, the
dose is 2-30 x 109 CFU. In another embodiment, the dose is 1-10 x 109 CFU. In
another
embodiment, the dose is 2-10 x 109 CFU. In another embodiment, the dose is 3-
10 x 109 CFU.
In another embodiment, the dose is 2-7 x 109 CFU. In another embodiment, the
dose is 2-5 x
109 CFU. In another embodiment, the dose is 3-5 x 109 CFU.
11001391In another embodiment, the dose is 1 x 109 organisms. In another
embodiment, the
dose is 1.5 x 109 organisms. In another embodiment, the dose is 2 x 109
organisms. In another
embodiment, the dose is 3 x 109 organisms. In another embodiment, the dose is
4 x 109
organisms. In another embodiment, the dose is 5 x 109 organisms. In another
embodiment, the
dose is 6 x 109 organisms. In another embodiment, the dose is 7 x 109
organisms. In another
embodiment, the dose is 8 x 109 organisms. In another embodiment, the dose is
10 x 109
organisms. In another embodiment, the dose is 1.5 x 1010 organisms. In another
embodiment,
32

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
the dose is 2 x 1010 organisms. In another embodiment, the dose is 2.5 x 1010
organisms. In
another embodiment, the dose is 3 x 1010 organisms. In another embodiment, the
dose is 3.3 x
1010 organisms. In another embodiment, the dose is 4 x 1010 organisms. In
another
embodiment, the dose is 5 x 1010 organisms.
1001401In another embodiment, the recombinant polypeptide of methods disclosed
herein is
expressed by the recombinant Listeria strain. In another embodiment, the
expression is
mediated by a nucleotide molecule carried by the recombinant Listeria strain.
1001411In another embodiment, the recombinant Listeria strain expresses the
recombinant
polypeptide by means of a plasmid that encodes the recombinant polypeptide. In
another
embodiment, the plasmid comprises a gene encoding a bacterial transcription
factor. In
another embodiment, the plasmid encodes a Listeria transcription factor. In
another
embodiment, the transcription factor is PrfA. In another embodiment, the PrfA
is a mutant
PrfA. In another embodiment, the PrfA mutant protein contains a D133V amino
acid
mutation.
1001421In another embodiment, the plasmid comprises a gene encoding a
metabolic enzyme.
In another embodiment, the metabolic enzyme is a bacterial metabolic enzyme.
In another
embodiment, the metabolic enzyme is a Listerial metabolic enzyme. In another
embodiment,
the metabolic enzyme is an amino acid metabolism enzyme. In another
embodiment, the
amino acid metabolism gene is involved in a cell wall synthesis pathway. In
another
embodiment, the metabolic enzyme is the product of a D-amino acid
aminotransferase gene
(dat). In another embodiment, the metabolic enzyme is the product of an
alanine racemase
gene (dal). In another embodiment, the metabolic enzyme is any other metabolic
enzyme
known in the art.
1001431In another embodiment, a method of present invention further comprises
the step of
boosting the human subject with a recombinant Listeria strain of the present
invention. In
another embodiment, the recombinant Listeria strain used in the booster
inoculation is the
same as the strain used in the initial "priming" inoculation. In another
embodiment, the
booster strain is different from the priming strain. In another embodiment,
the same doses are
used in the priming and boosting inoculations. In another embodiment, a larger
dose is used in
the booster. In another embodiment, a smaller dose is used in the booster.
1001441In another embodiment, a method of present invention further comprises
the step of
inoculating the human subject with an immunogenic composition comprising the
E7 antigen.
In another embodiment, the immunogenic composition comprises a recombinant E7
protein or
fragment thereof. In another embodiment, the immunogenic composition comprises
a
33

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
nucleotide molecule expressing a recombinant E7 protein or fragment thereof.
In another
embodiment, the non-Listerial inoculation is administered after the Listerial
inoculation. In
another embodiment, the non-Listerial inoculation is administered before the
Listerial
inoculation.
1001451"Boosting" refers, in another embodiment, to administration of an
additional vaccine
dose to a subject. In another embodiment of methods of the present invention,
2 boosts (or a
total of 3 inoculations) are administered. In another embodiment, 3 boosts are
administered.
In another embodiment, 4 boosts are administered. In another embodiment, 5
boosts are
administered. In another embodiment, 6 boosts are administered. In another
embodiment,
more than 6 boosts are administered.
1001461The recombinant Listeria strain of methods and compositions disclosed
herein is, in
another embodiment, a recombinant Listeria monocytogenes strain. In another
embodiment,
the Listeria strain is a recombinant Listeria seeligeri strain. In another
embodiment, the
Listeria strain is a recombinant Listeria grayi strain. In another embodiment,
the Listeria
strain is a recombinant Listeria ivanovii strain. In another embodiment, the
Listeria strain is a
recombinant Listeria murrayi strain. In another embodiment, the Listeria
strain is a
recombinant Listeria welshimeri strain. In another embodiment, the Listeria
strain is a
recombinant strain of any other Listeria species known in the art.
1001471The present invention provides a number of listerial species and
strains for making or
engineering an attenuated Listeria of the present invention. In one
embodiment, the Listeria
strain is L. monocytogenes 10403S wild type (see Bishop and Hinrichs (1987) J.
Immunol.
139: 2005-2009; Lauer, et al. (2002) J. Bact. 184: 4177-4186.) In another
embodiment, the
Listeria strain is L. monocytogenes DP-L4056 (phage cured) (see Lauer, et al.
(2002) J. Bact.
184: 4177-4186). In another embodiment, the Listeria strain is L.
monocytogenes DP-L4027,
which is phage cured and deleted in the hly gene (see Lauer, et al. (2002) J.
Bact. 184: 4177-
4186; Jones and Portnoy (1994) Infect. Immunity 65: 5608-5613.). In another
embodiment,
the Listeria strain is L. monocytogenes DP-L4029, which is phage cured,
deleted in ActA
(see Lauer, et al. (2002) J. Bact. 184: 4177-4186; Skoble, et al. (2000) J.
Cell Biol. 150: 527-
538). In another embodiment, the Listeria strain is L. monocytogenes DP-L4042
(delta PEST)
(see Brockstedt, et al. (2004) Proc. Natl. Acad. Sci. USA 101: 13832-13837;
supporting
information). In another embodiment, the Listeria strain is L monocyto genes
DP-L4097
(LLO-544A) (see Brockstedt, et al. (2004) Proc. Natl. Acad. Sci. USA 101:
13832-13837;
supporting information). In another embodiment, the Listeria strain is L.
monocytogenes DP-
L4364 (delta 1p1A; lipoate protein ligase) (see Brockstedt, et al. (2004)
Proc. Natl. Acad. Sci.
34

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
USA 101: 13832-13837; supporting information). In another embodiment, the
Listeria strain
is L. monocytogenes DP-L4405 (delta in1A) (see Brockstedt, et al. (2004) Proc.
Natl. Acad.
Sci. USA 101: 13832-13837; supporting information). In another embodiment, the
Listeria
strain is L. monocytogenes DP-L4406 (delta in1B) (see Brockstedt, et al.
(2004) Proc. Natl.
Acad. Sci. USA 101: 13832-13837; supporting information). In another
embodiment, the
Listeria strain is L. monocytogenes CS-L0001 (delta ActA-delta in1B) (see
Brockstedt, et al.
(2004) Proc. Natl. Acad. Sci. USA 101: 13832-13837; supporting information).
In another
embodiment, the Listeria strain is L. monocytogenes CS-L0002 (delta ActA-delta
1p1A) (see
Brockstedt, et al. (2004) Proc. Natl. Acad. Sci. USA 101: 13832-13837;
supporting
information). In another embodiment, the Listeria strain is L. monocytogenes
CS-L0003
(L461T-delta 1p1A) (see Brockstedt, et al. (2004) Proc. Natl. Acad. Sci. USA
101: 13832-
13837; supporting information). In another embodiment, the Listeria strain is
L.
monocytogenes DP-L4038 (delta ActA-LLO L461T) (see Brockstedt, et al. (2004)
Proc. Natl.
Acad. Sci. USA 101: 13832-13837; supporting information). In another
embodiment, the
Listeria strain is L. monocytogenes DP-L4384 (544A-LLO L461T) (see Brockstedt,
et al.
(2004) Proc. Natl. Acad. Sci. USA 101: 13832-13837; supporting information).
In another
embodiment, the Listeria strain is L. monocyto genes. Mutation in lipoate
protein (see
O'Riordan, et al. (2003) Science 302: 462-464). In another embodiment, the
Listeria strain is
L. monocytogenes DP-L4017 (10403S hly (L461T), having a point mutation in
hemolysin
gene (see U.S. Provisional Pat. Appl. Ser. No. 60/490,089 filed Jul. 24,
2003). In another
embodiment, the Listeria strain is L. monocytogenes EGD (see GenBank Acc. No.
AL591824). In another embodiment, the Listeria strain is L. monocytogenes EGD-
e (see
GenBank Acc. No. NC_003210. ATCC Acc. No. BAA-679). In another embodiment, the

Listeria strain is L. monocytogenes DP-L4029 deleted in uvrAB (see U.S.
Provisional Pat.
Appl. Ser. No. 60/541,515 filed Feb. 2, 2004; US Provisional Pat. Appl. Ser.
No. 60/490,080
filed Jul. 24, 2003). In another embodiment, the Listeria strain is L.
monocytogenes ActA-
/in1B - double mutant (see ATCC Acc. No. PTA-5562). In another embodiment, the
Listeria
strain is L. monocytogenes lplA mutant or hly mutant (see U.S. Pat. Applic.
No. 20040013690
of Portnoy, et. al). In another embodiment, the Listeria strain is L.
monocytogenes DAL/DAT
double mutant. (see U.S. Pat. Applic. No. 20050048081 of Frankel and Portnoy.
The present
invention encompasses reagents and methods that comprise the above listerial
strains, as well
as these strains that are modified, e.g., by a plasmid and/or by genomic
integration, to contain
a nucleic acid encoding one of, or any combination of, the following genes:
hly (LLO;
listeriolysin); iap (p60); in1A; in1B; in1C; dal (alanine racemase); dat (D-
amino acid

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
aminotransferase); plcA; plcB; actA; or any nucleic acid that mediates growth,
spread,
breakdown of a single walled vesicle, breakdown of a double walled vesicle,
binding to a host
cell, uptake by a host cell. The present invention is not to be limited by the
particular strains
disclosed above.
1001481In another embodiment, a recombinant Listeria strain disclosed herein
has been
passaged through an animal host. In another embodiment, the passaging
maximizes efficacy
of the strain as a vaccine vector. In another embodiment, the passaging
stabilizes the
immunogenicity of the Listeria strain. In another embodiment, the passaging
stabilizes the
virulence of the Listeria strain. In another embodiment, the passaging
increases the
immunogenicity of the Listeria strain. In another embodiment, the passaging
increases the
virulence of the Listeria strain. In another embodiment, the passaging removes
unstable sub-
strains of the Listeria strain. In another embodiment, the passaging reduces
the prevalence of
unstable sub-strains of the Listeria strain. In another embodiment, the
Listeria strain contains
a genomic insertion of the gene encoding the antigen-containing recombinant
peptide. In
another embodiment, the Listeria strain carries a plasmid comprising the gene
encoding the
antigen-containing recombinant peptide. In another embodiment, the passaging
is performed
as described herein (e.g. in Example 12). In another embodiment, the passaging
is performed
by any other method known in the art.
1001491In another embodiment, the recombinant Listeria strain utilized in
methods disclosed
herein has been stored in a frozen cell bank. In another embodiment, the
recombinant Listeria
strain has been stored in a lyophilized cell bank.
1001501In another embodiment, the cell bank of methods and compositions
disclosed herein is
a master cell bank. In another embodiment, the cell bank is a working cell
bank. In another
embodiment, the cell bank is Good Manufacturing Practice (GMP) cell bank. In
another
embodiment, the cell bank is intended for production of clinical-grade
material. In another
embodiment, the cell bank conforms to regulatory practices for human use. In
another
embodiment, the cell bank is any other type of cell bank known in the art.
1001511"Good Manufacturing Practices" are defined, in another embodiment, by
(21 CFR
210-211) of the United States Code of Federal Regulations. In another
embodiment, "Good
Manufacturing Practices" are defined by other standards for production of
clinical-grade
material or for human consumption; e.g. standards of a country other than the
United States.
1001521In another embodiment, a recombinant Listeria strain utilized in
methods disclosed
herein is from a batch of vaccine doses.
36

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
1001531In another embodiment, a recombinant Listeria strain utilized in
methods disclosed
herein is from a frozen or lyophilized stock produced by methods provided in
US Patent Ser.
No. 8,114,414, which is incorporated by reference herein.
1001541In another embodiment, a peptide disclosed herein is a fusion peptide.
In another
embodiment, "fusion peptide" refers to a peptide or polypeptide comprising 2
or more
proteins linked together by peptide bonds or other chemical bonds. In another
embodiment,
the proteins are linked together directly by a peptide or other chemical bond.
In another
embodiment, the proteins are linked together with 1 or more AA (e.g. a
"spacer") between the
2 or more proteins.
1001551In another embodiment, a vaccine disclosed herein further comprises an
adjuvant. The
adjuvant utilized in methods and compositions disclosed herein is, in another
embodiment, a
granulocyte/macrophage colony-stimulating factor (GM-C SF) protein. In another

embodiment, the adjuvant comprises a GM-CSF protein. In another embodiment,
the adjuvant
is a nucleotide molecule encoding GM-CSF. In another embodiment, the adjuvant
comprises
a nucleotide molecule encoding GM-CSF. In another embodiment, the adjuvant is
saponin
Q521. In another embodiment, the adjuvant comprises saponin Q521. In another
embodiment, the adjuvant is monophosphoryl lipid A. In another embodiment, the
adjuvant
comprises monophosphoryl lipid A. In another embodiment, the adjuvant is
SBAS2. In
another embodiment, the adjuvant comprises SBAS2. In another embodiment, the
adjuvant is
an unmethylated CpG-containing oligonucleotide. In another embodiment, the
adjuvant
comprises an unmethylated CpG-containing oligonucleotide. In another
embodiment, the
adjuvant is an immune-stimulating cytokine. In another embodiment, the
adjuvant comprises
an immune-stimulating cytokine. In another embodiment, the adjuvant is a
nucleotide
molecule encoding an immune-stimulating cytokine. In another embodiment, the
adjuvant
comprises a nucleotide molecule encoding an immune-stimulating cytokine. In
another
embodiment, the adjuvant is or comprises a quill glycoside. In another
embodiment, the
adjuvant is or comprises a bacterial mitogen. In another embodiment, the
adjuvant is or
comprises a bacterial toxin. In another embodiment, the adjuvant is or
comprises any other
adjuvant known in the art.
1001561In another embodiment, a nucleotide disclosed herein is operably linked
to a
promoter/regulatory sequence that drives expression of the encoded peptide in
the Listeria
strain. Promoter/regulatory sequences useful for driving constitutive
expression of a gene are
well known in the art and include, but are not limited to, for example, the
PhlyA, PActA, and p60
promoters of Listeria, the Streptococcus bac promoter, the Streptomyces
griseus sgiA
37

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
promoter, and the B. thuringiensis phaZ promoter. In another embodiment,
inducible and
tissue specific expression of the nucleic acid encoding a peptide disclosed
herein is
accomplished by placing the nucleic acid encoding the peptide under the
control of an
inducible or tissue specific promoter/regulatory sequence. Examples of tissue
specific or
inducible promoter/regulatory sequences which are useful for his purpose
include, but are not
limited to the MMTV LTR inducible promoter, and the SV40 late
enhancer/promoter. In
another embodiment, a promoter that is induced in response to inducing agents
such as
metals, glucocorticoids, and the like, is utilized. Thus, it will be
appreciated that the invention
includes the use of any promoter/regulatory sequence, which is either known or
unknown, and
which is capable of driving expression of the desired protein operably linked
thereto.
1001571An N-terminal fragment of an ActA protein utilized in methods and
compositions
disclosed herein has, in another embodiment, the sequence set forth in SEQ ID
NO: 5:
MRAMMVVFITANCITINPDHFAATDSEDSSLNTDEWEEEKTEEQPSEVNTGPR
YETAREVSSRDIKELEKSNKVRNTNKADLIAMLKEKAEKGPNINNNNSEQTENAA
INEEASGADRPAIQVERRHPGLPSDSAAEIKKRRKAIAS SDSELESLTYPDKPTKVN
KKKVAKESVADASESDLDSSMQSADESSPQPLKANQQPFFPKVFKKIKDAGKWV
RDKIDENPEVKKAIVDKSAGLIDQLLTKKKSEEVNASDFPPPPTDEELRLALPETP
MLLGFNAPATSEPSSFEFPPPPTDEELRLALPETPMLLGFNAPATSEPSSFEFPPPPT
EDELEIIRETAS SLDSSFTRGDLASLRNAINRHSQNFSDFPPIPTEEELNGRGGRP. In
another embodiment, the ActA fragment comprises the sequence set forth in SEQ
ID NO:
5. In another embodiment, the ActA fragment is any other ActA fragment known
in the
art.
1001581In another embodiment, the recombinant nucleotide encoding a fragment
of an ActA
protein comprises the sequence set forth in SEQ ID NO: 6:
Atgcgtgcgatgatggtggttttcattactgccaattgcattacgattaaccccgacataatatttgcagcgacagata
gcgaa
gattctagtctaaacacagatgaatgggaagaagaaaaaacagaagagcaaccaagcgaggtaaatacgggaccaagat
acg
aaactgcacgtgaagtaagttcacgtgatattaaagaactagaaaaatcgaataaagtgagaaatacgaacaaagcaga
cctaat
agcaatgttgaaagaaaaagcagaaaaaggtccaaatatcaataataacaacagtgaacaaactgagaatgcggctata
aatga
agaggettcaggagccgaccgaccagctatacaagtggagcgtcgtcatccaggattgccatcggatagcgcageggaa
atta
aaaaaagaaggaaagccatagcatcatcggatagtgagcttgaaagccttacttatccggataaaccaacaaaagtaaa
taagaa
aaaagtggcgaaagagtcagttgeggatgatctgaaagtgacttagattctagcatgcagtcagcagatgagtatcacc
acaac
ctttaaaagcaaaccaacaaccatttttccctaaagtatttaaaaaaataaaagatgeggggaaatgggtacgtgataa
aatcgacg
aaaatcctgaagtaaagaaagcgattgttgataaaagtgcagggttaattgaccaattattaaccaaaaagaaaagtga
agaggta
aatgatcggacttcccgccaccacctacggatgaagagttaagacttgetttgccagagacaccaatgcncttggtttt
aatgctc
38

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
ctgctacatcagaaccgagctcattcgaatttccaccaccacctacggatgaagagttaagacttgctttgccagagac
gcc aatg
cttcttggttttaatgctcctgctac atcggaaccgagctc gttcgaatttcc accgcctcc
aacagaagatgaactagaaatc atcc
gggaaacagcatcctcgctagattctagttttac aagaggggatttagctagtttgagaaatgctattaatcgcc
atagtc aaaatttc
tctgatttcccaccaatcccaacagaagaagagttgaacgggagaggcggtagacca. In another
embodiment, the
recombinant nucleotide has the sequence set forth in SEQ ID NO: 6. In another
embodiment, the recombinant nucleotide comprises any other sequence that
encodes a
fragment of an ActA protein.
1001591In another embodiment of the methods and compositions of the present
invention, a
PEST amino acid AA sequence is fused to the E7 or E6 antigen. As disclosed
herein,
recombinant Listeria strains expressing PEST amino acid sequence-antigen
fusions induce
anti-tumor immunity (Example 3) and generate antigen-specific, tumor-
infiltrating T cells
(Example 4). Further, enhanced cell mediated immunity was demonstrated for
fusion proteins
comprising an antigen and LLO containing the PEST amino acid AA sequence
KENSISSMAPPASPPASPKTPIEKKHADEIDK (SEQ ID NO: 1).
1001601Thus, fusion of an antigen to other LM PEST amino acid sequences and
PEST amino
acid sequences derived from other prokaryotic organisms will also enhance
immunogenicity
of the antigen. The PEST amino acid AA sequence has, in another embodiment, a
sequence
selected from SEQ ID NO: 7-12. In another embodiment, the PEST amino acid
sequence is a
PEST amino acid sequence from the LM ActA protein. In another embodiment, the
PEST
amino acid sequence is KTEEQPSEVNTGPR (SEQ ID NO: 7),
KASVTDTSEGDLDSSMQSADESTPQPLK (SEQ ID NO: 8),
KNEEVNASDFPPPPTDEELR (SEQ ID NO: 9), or
RGGIPTSEEFSSLNSGDFTDDENSETTEEEIDR (SEQ ID NO: 10). In another embodiment,
the PEST amino acid sequence is from Streptolysin 0 protein of Streptococcus
sp. In another
embodiment, the PEST amino acid sequence is from Streptococcus pyogenes
Streptolysin 0,
e.g. KQNTASTETTTTNEQPK (SEQ ID NO: 11) at AA 35-51. In another embodiment, the

PEST amino acid sequence is from Streptococcus equisimilis Streptolysin 0,
e.g.
KQNTANTETTTTNEQPK (SEQ ID NO:12) at AA 38-54. In another embodiment, the PEST
amino acid sequence is another PEST amino acid AA sequence derived from a
prokaryotic
organism. In another embodiment, the PEST amino acid sequence is any other
PEST amino
acid sequence known in the art.
1001611PEST amino acid sequences of other prokaryotic organism can be
identified in
accordance with methods such as described by, for example Rechsteiner and
Rogers (1996,
Trends Biochem. Sci. 21:267-271) for LM. Alternatively, PEST amino acid AA
sequences
39

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
from other prokaryotic organisms can also be identified based by this method.
Other
prokaryotic organisms wherein PEST amino acid AA sequences would be expected
to
include, but are not limited to, other Listeria species. In another
embodiment, the PEST amino
acid sequence is embedded within the antigenic protein. Thus, in another
embodiment,
"fusion" refers to an antigenic protein comprising both the antigen and the
PEST amino acid
amino acid sequence either linked at one end of the antigen or embedded within
the antigen.
11001621 In another embodiment, the PEST amino acid sequence is identified
using any other
method or algorithm known in the art, e.g the CaSPredictor (Garay-Malpartida
HM,
Occhiucci JM, Alves J, Belizario JE. Bioinformatics. 2005 Jun;21 Suppl 1:i169-
76). In
another embodiment, the following method is used:
[00163[A PEST index is calculated for each 30-35 AA stretch by assigning a
value of 1 to the
amino acids Ser, Thr, Pro, Glu, Asp, Asn, or Gln. The coefficient value (CV)
for each of the
PEST residue is 1 and for each of the other AA (non-PEST) is 0.
[00164]
11001651 In another embodiment, the LLO protein, ActA protein, or fragment
thereof disclosed
herein need not be that which is set forth exactly in the sequences set forth
herein, but rather
other alterations, modifications, or changes can be made that retain the
functional
characteristics of an LLO or ActA protein fused to an antigen as set forth
elsewhere herein. In
another embodiment, the present invention utilizes an analog of an LLO
protein, ActA
protein, or fragment thereof. Analogs differ, in another embodiment, from
naturally occurring
proteins or peptides by conservative AA sequence differences or by
modifications which do
not affect sequence, or by both.
11001661 In another embodiment, either a whole E7 protein or a fragment
thereof is fused to a
LLO protein, ActA protein, or PEST amino acid sequence-containing peptide to
generate a
recombinant peptide of methods of the present invention. The E7 protein that
is utilized
(either whole or as the source of the fragments) has, in another embodiment,
the sequence
[00167] MHGDTPTLHEYMLDLQPETTDLYCYEQLND S S EEEDEID GPAGQAEPDRAHY
NIVTFCCKCDSTLRLCVQSTHVDIRTLEDLLMGTLGIVCPICSQKP (SEQ ID No: 13). In
another embodiment, the E7 protein is a homologue of SEQ ID No: 13. In another
embodiment, the E7 protein is a variant of SEQ ID No: 13. In another
embodiment, the E7
protein is an isomer of SEQ ID No: 13. In another embodiment, the E7 protein
is a fragment
of SEQ ID No: 13. In another embodiment, the E7 protein is a fragment of a
homologue of
SEQ ID No: 13. In another embodiment, the E7 protein is a fragment of a
variant of SEQ ID
No: 13. In another embodiment, the E7 protein is a fragment of an isomer of
SEQ ID No: 13.

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[00168]In another embodiment, the sequence of the E7 protein is:
[00169] MHGPKATLQDIVLHLEPQNEIPVDLLCHEQLS DS EEENDEID GVNHQHLPARR
AEPQRHTMLC M C C KC EARIELVVES S ADD LRAFQQLFLNTLSFVCPWC AS QQ (SEQ
ID No: 14). In another embodiment, the E6 protein is a homologue of SEQ ID No:
14. In
another embodiment, the E6 protein is a variant of SEQ ID No: 14. In another
embodiment,
the E6 protein is an isomer of SEQ ID No: 14. In another embodiment, the E6
protein is a
fragment of SEQ ID No: 14. In another embodiment, the E6 protein is a fragment
of a
homologue of SEQ ID No: 14. In another embodiment, the E6 protein is a
fragment of a
variant of SEQ ID No: 14. In another embodiment, the E6 protein is a fragment
of an isomer
of SEQ ID No: 14.
[00170]In another embodiment, the E7 protein has a sequence set forth in one
of the
following GenBank entries: M24215, NC_004500, V01116, X62843, or M14119. In
another
embodiment, the E7 protein is a homologue of a sequence from one of the above
GenBank
entries. In another embodiment, the E7 protein is a variant of a sequence from
one of the
above GenBank entries. In another embodiment, the E7 protein is an isomer of a
sequence
from one of the above GenBank entries. In another embodiment, the E7 protein
is a fragment
of a sequence from one of the above GenBank entries. In another embodiment,
the E7 protein
is a fragment of a homologue of a sequence from one of the above GenBank
entries. In
another embodiment, the E7 protein is a fragment of a variant of a sequence
from one of the
above GenBank entries. In another embodiment, the E7 protein is a fragment of
an isomer of
a sequence from one of the above GenBank entries..
11001711 In another embodiment, either a whole E6 protein or a fragment
thereof is fused to a
LLO protein, ActA protein, or PEST amino acid sequence-containing peptide to
generate a
recombinant peptide of methods of the present invention. The E6 protein that
is utilized
(either whole or as the source of the fragments) has, in another embodiment,
the sequence
[00172] MHQKRTAMFQDPQERPRKLPQLCTELQTTIHDIILECVYC KQQLLRREVYDFA
FRDLC IVYRDGNPYAVCD KC LKFYS KIS EYRHYCYS LYGTTLEQQYNKPLC DLLIRCI
NCQKPLCPEEKQRHLDKKQRFHNIRGRWTGRCMSCCRSSRTRRETQL (SEQ ID No:
15). In another embodiment, the E6 protein is a homologue of SEQ ID No: 15. In
another
embodiment, the E6 protein is a variant of SEQ ID No: 15. In another
embodiment, the E6
protein is an isomer of SEQ ID No: 15. In another embodiment, the E6 protein
is a fragment
of SEQ ID No: 15. In another embodiment, the E6 protein is a fragment of a
homologue of
SEQ ID No: 15. In another embodiment, the E6 protein is a fragment of a
variant of SEQ ID
No: 15. In another embodiment, the E6 protein is a fragment of an isomer of
SEQ ID No: 15.
41

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[00173]In another embodiment, the sequence of the E6 protein is:
[00174] MARFEDPTRRPYKLPDLC TELNTS LQDIEIT CVYC KTVLELTEVFEFAFKD LFV
VYRDSIPHAACHKCIDFYSRIRELRHYSDSVYGDTLEKLTNTGLYNLLIRCLRCQKPL
NPAEKLRHLNEKRRFHNIAGHYRGQCHSCCNRARQERLQRRRETQV (SEQ ID No:
16). In another embodiment, the E6 protein is a homologue of SEQ ID No: 16. In
another
embodiment, the E6 protein is a variant of SEQ ID No: 16. In another
embodiment, the E6
protein is an isomer of SEQ ID No: 16. In another embodiment, the E6 protein
is a fragment
of SEQ ID No: 16. In another embodiment, the E6 protein is a fragment of a
homologue of
SEQ ID No: 16. In another embodiment, the E6 protein is a fragment of a
variant of SEQ ID
No: 16. In another embodiment, the E6 protein is a fragment of an isomer of
SEQ ID No: 16.
[00175]In another embodiment, the E6 protein has a sequence set forth in one
of the
following GenBank entries: M24215, M14119, NC_004500, V01116, X62843, or
M14119. In
another embodiment, the E6 protein is a homologue of a sequence from one of
the above
GenBank entries. In another embodiment, the E6 protein is a variant of a
sequence from one
of the above GenBank entries. In another embodiment, the E6 protein is an
isomer of a
sequence from one of the above GenBank entries. In another embodiment, the E6
protein is a
fragment of a sequence from one of the above GenBank entries. In another
embodiment, the
E6 protein is a fragment of a homologue of a sequence from one of the above
GenBank
entries. In another embodiment, the E6 protein is a fragment of a variant of a
sequence from
one of the above GenBank entries. In another embodiment, the E6 protein is a
fragment of an
isomer of a sequence from one of the above GenBank entries.
[00176]In another embodiment, "homology" refers to identity to an LLO sequence
(e.g. to
one of SEQ ID No: 2-4) of greater than 70%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 2-4 of greater than 64%. In another embodiment,
"homology"
refers to identity to one of SEQ ID No: 2-4 of greater than 68%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 2-4 of greater than 72%. In
another
embodiment, "homology" refers to identity to one of SEQ ID No: 2-4 of greater
than 75%. In
another embodiment, "homology" refers to identity to one of SEQ ID No: 2-4 of
greater than
78%. In another embodiment, "homology" refers to identity to one of SEQ ID No:
2-4 of
greater than 80%. In another embodiment, "homology" refers to identity to one
of SEQ ID
No: 2-4 of greater than 82%. In another embodiment, "homology" refers to
identity to one of
SEQ ID No: 2-4 of greater than 83%. In another embodiment, "homology" refers
to identity
to one of SEQ ID No: 2-4 of greater than 85%. In another embodiment,
"homology" refers to
identity to one of SEQ ID No: 2-4 of greater than 87%. In another embodiment,
"homology"
42

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
refers to identity to one of SEQ ID No: 2-4 of greater than 88%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 2-4 of greater than 90%. In
another
embodiment, "homology" refers to identity to one of SEQ ID No: 2-4 of greater
than 92%. In
another embodiment, "homology" refers to identity to one of SEQ ID No: 2-4 of
greater than
93%. In another embodiment, "homology" refers to identity to one of SEQ ID No:
2-4 of
greater than 95%. In another embodiment, "homology" refers to identity to one
of SEQ ID
No: 2-4 of greater than 96%. In another embodiment, "homology" refers to
identity to one of
SEQ ID No: 2-4 of greater than 97%. In another embodiment, "homology" refers
to identity
to one of SEQ ID No: 2-4 of greater than 98%. In another embodiment,
"homology" refers to
identity to one of SEQ ID No: 2-4 of greater than 99%. In another embodiment,
"homology"
refers to identity to one of SEQ ID No: 2-4 of 100%.
1001771In another embodiment, "homology" refers to identity to an E7 sequence
(e.g. to one
of SEQ ID No: 13-14) of greater than 70%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 13-14 of greater than 62%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 13-14 of greater than 64%.
In another
embodiment, "homology" refers to identity to one of SEQ ID No: 13-14 of
greater than 68%.
In another embodiment, "homology" refers to identity to one of SEQ ID No: 13-
14 of greater
than 72%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 13-14
of greater than 75%. In another embodiment, "homology" refers to identity to
one of SEQ ID
No: 13-14 of greater than 78%. In another embodiment, "homology" refers to
identity to one
of SEQ ID No: 13-14 of greater than 80%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 13-14 of greater than 82%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 13-14 of greater than 83%.
In another
embodiment, "homology" refers to identity to one of SEQ ID No: 13-14 of
greater than 85%.
In another embodiment, "homology" refers to identity to one of SEQ ID No: 13-
14 of greater
than 87%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 13-14
of greater than 88%. In another embodiment, "homology" refers to identity to
one of SEQ ID
No: 13-14 of greater than 90%. In another embodiment, "homology" refers to
identity to one
of SEQ ID No: 13-14 of greater than 92%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 13-14 of greater than 93%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 13-14 of greater than 95%.
In another
embodiment, "homology" refers to identity to one of SEQ ID No: 13-14 of
greater than 96%.
In another embodiment, "homology" refers to identity to one of SEQ ID No: 13-
14 of greater
than 97%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 13-14
43

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
of greater than 98%. In another embodiment, "homology" refers to identity to
one of SEQ ID
No: 13-14 of greater than 99%. In another embodiment, "homology" refers to
identity to one
of SEQ ID No: 13-14 of 100%.
[001781In another embodiment, "homology" refers to identity to an E6 sequence
(e.g. to one
of SEQ ID No: 15-16) of greater than 70%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 15-16 of greater than 64%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 15-16 of greater than 68%.
In another
embodiment, "homology" refers to identity to one of SEQ ID No: 15-16 of
greater than 72%.
In another embodiment, "homology" refers to identity to one of SEQ ID No: 15-
16 of greater
than 75%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 15-16
of greater than 78%. In another embodiment, "homology" refers to identity to
one of SEQ ID
No: 15-16 of greater than 80%. In another embodiment, "homology" refers to
identity to one
of SEQ ID No: 15-16 of greater than 82%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 15-16 of greater than 83%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 15-16 of greater than 85%.
In another
embodiment, "homology" refers to identity to one of SEQ ID No: 15-16 of
greater than 87%.
In another embodiment, "homology" refers to identity to one of SEQ ID No: 15-
16 of greater
than 88%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 15-16
of greater than 90%. In another embodiment, "homology" refers to identity to
one of SEQ ID
No: 15-16 of greater than 92%. In another embodiment, "homology" refers to
identity to one
of SEQ ID No: 15-16 of greater than 93%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 15-16 of greater than 95%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 15-16 of greater than 96%.
In another
embodiment, "homology" refers to identity to one of SEQ ID No: 15-16 of
greater than 97%.
In another embodiment, "homology" refers to identity to one of SEQ ID No: 15-
16 of greater
than 98%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 15-16
of greater than 99%. In another embodiment, "homology" refers to identity to
one of SEQ ID
No: 15-16 of 100%.
[001791In another embodiment, "homology" refers to identity to a PEST amino
acid sequence
(e.g. to one of SEQ ID No: 1, and 7-12) or to an ActA sequence (e.g. to one of
SEQ ID No: 5-
6) of greater than 70%. In another embodiment, "homology" refers to identity
to one of SEQ
ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than 60%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-
6 of greater
than 64%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 1, and
44

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
7-12 or SEQ ID No: 5-6 of greater than 68%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than
72%. In another
embodiment, "homology" refers to identity to one of SEQ ID No: 1, and 7-12 or
SEQ ID No:
5-6 of greater than 75%. In another embodiment, "homology" refers to identity
to one of SEQ
ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than 78%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-
6 of greater
than 80%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 1, and
7-12 or SEQ ID No: 5-6 of greater than 82%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than
83%. In another
embodiment, "homology" refers to identity to one of SEQ ID No: 1, and 7-12 or
SEQ ID No:
5-6 of greater than 85%. In another embodiment, "homology" refers to identity
to one of SEQ
ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than 87%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-
6 of greater
than 88%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 1, and
7-12 or SEQ ID No: 5-6 of greater than 90%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than
92%. In another
embodiment, "homology" refers to identity to one of SEQ ID No: 1, and 7-12 or
SEQ ID No:
5-6 of greater than 93%. In another embodiment, "homology" refers to identity
to one of SEQ
ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than 95%. In another
embodiment,
"homology" refers to identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-
6 of greater
than 96%. In another embodiment, "homology" refers to identity to one of SEQ
ID No: 1, and
7-12 or SEQ ID No: 5-6 of greater than 97%. In another embodiment, "homology"
refers to
identity to one of SEQ ID No: 1, and 7-12 or SEQ ID No: 5-6 of greater than
98%. In another
embodiment, "homology" refers to identity to one of SEQ ID No: 1, and 7-12 or
SEQ ID No:
5-6 of greater than 99%. In another embodiment, "homology" refers to identity
to one of SEQ
ID No: 1, and 7-12 or SEQ ID No: 5-6 of 100%.
[001801Protein and/or peptide homology for any AA sequence listed herein is
determined, in
one embodiment, by methods well described in the art, including immunoblot
analysis, or via
computer algorithm analysis of AA sequences, utilizing any of a number of
software packages
available, via established methods. Some of these packages include the FASTA,
BLAST,
MPsrch or Scanps packages, and employ, in other embodiments, the use of the
Smith and
Waterman algorithms, and/or global/local or BLOCKS alignments for analysis,
for example.
[001811In another embodiment, the LLO protein, ActA protein, or fragment
thereof is
attached to the antigen by chemical conjugation. In another embodiment,
glutaraldehyde is

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
used for the conjugation. In another embodiment, the conjugation is performed
using any
suitable method known in the art.
1001821In another embodiment, fusion proteins disclosed herein are prepared by
any suitable
method, including, for example, cloning and restriction of appropriate
sequences or direct
chemical synthesis by methods discussed below. In another embodiment,
subsequences are
cloned and the appropriate subsequences cleaved using appropriate restriction
enzymes. The
fragments are then ligated, in another embodiment, to produce the desired DNA
sequence. In
another embodiment, DNA encoding the fusion protein is produced using DNA
amplification
methods, for example polymerase chain reaction (PCR). First, the segments of
the native
DNA on either side of the new terminus are amplified separately. The 5 end of
the one
amplified sequence encodes the peptide linker, while the 3' end of the other
amplified
sequence also encodes the peptide linker. Since the 5' end of the first
fragment is
complementary to the 3' end of the second fragment, the two fragments (after
partial
purification, e.g. on LMP agarose) can be used as an overlapping template in a
third PCR
reaction. The amplified sequence will contain codons, the segment on the
carboxy side of the
opening site (now forming the amino sequence), the linker, and the sequence on
the amino
side of the opening site (now forming the carboxyl sequence). The insert is
then ligated into a
plasmid.
1001831In another embodiment, the LLO protein, ActA protein, or fragment
thereof and the
antigen, or fragment thereof are conjugated by a means known to those of skill
in the art. In
another embodiment, the antigen, or fragment thereof is conjugated, either
directly or through
a linker (spacer), to the ActA protein or LLO protein. In another embodiment,
the chimeric
molecule is recombinantly expressed as a single-chain fusion protein.
1001841In another embodiment, a fusion peptide disclosed herein is synthesized
using
standard chemical peptide synthesis techniques. In another embodiment, the
chimeric
molecule is synthesized as a single contiguous polypeptide. In another
embodiment, the LLO
protein, ActA protein, or fragment thereof; and the antigen, or fragment
thereof are
synthesized separately, then fused by condensation of the amino terminus of
one molecule
with the carboxyl terminus of the other molecule, thereby forming a peptide
bond. In another
embodiment, the ActA protein or LLO protein and antigen are each condensed
with one end
of a peptide spacer molecule, thereby forming a contiguous fusion protein.
1001851In another embodiment, the peptides and proteins disclosed herein are
prepared by
solid-phase peptide synthesis (SPPS) as described by Stewart et al. in Solid
Phase Peptide
Synthesis, 2nd Edition, 1984, Pierce Chemical Company, Rockford, Ill.; or as
described by
46

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
Bodanszky and Bodanszky (The Practice of Peptide Synthesis, 1984, Springer-
Verlag, New
York). In another embodiment, a suitably protected AA residue is attached
through its
carboxyl group to a derivatized, insoluble polymeric support, such as cross-
linked polystyrene
or polyamide resin. "Suitably protected" refers to the presence of protecting
groups on both
the alpha-amino group of the amino acid, and on any side chain functional
groups. Side chain
protecting groups are generally stable to the solvents, reagents and reaction
conditions used
throughout the synthesis, and are removable under conditions which will not
affect the final
peptide product. Stepwise synthesis of the oligopeptide is carried out by the
removal of the N-
protecting group from the initial AA, and couple thereto of the carboxyl end
of the next AA in
the sequence of the desired peptide. This AA is also suitably protected. The
carboxyl of the
incoming AA can be activated to react with the N-terminus of the support-bound
AA by
formation into a reactive group such as formation into a carbodiimide, a
symmetric acid
anhydride or an "active ester" group such as hydroxybenzotriazole or
pentafluorophenly
esters.
11001861 In another embodiment, the present invention provides a kit
comprising vaccine of the
present invention, an applicator, and instructional material that describes
use of the methods
of the invention. Although model kits are described below, the contents of
other useful kits
will be apparent to the skilled artisan in light of the present disclosure.
EXPERIMENTAL DETAILS SECTION
EXAMPLE 1: LLO-ANTIGEN FUSIONS INDUCE ANTI-TUMOR IMMUNITY
MATERIALS AND EXPERIMENTAL METHODS (EXAMPLES 1-2)
Cell lines
[00187] The C57BL/6 syngeneic TC-1 tumor was immortalized with HPV-16 E6 and
E7 and
transformed with the c-Ha-ras oncogene. TC-1, provided by T. C. Wu (Johns
Hopkins
University School of Medicine, Baltimore, MD) is a highly tumorigenic lung
epithelial cell
expressing low levels of with HPV-16 E6 and E7 and transformed with the c-Ha-
ras
oncogene. TC-1 was grown in RPMI 1640, 10% FCS, 2 mM L-glutamine, 100 U/ml
penicillin, 100 p g/ml streptomycin, 100 p M nonessential amino acids, 1 mM
sodium
pyruvate, 50 micromolar (mcM) 2-ME, 400 microgram (mcg)/m1 G418, and 10%
National
Collection Type Culture-109 medium at 37 with 10% CO2. C3 is a mouse embryo
cell from
C57BL/6 mice immortalized with the complete genome of HPV 16 and transformed
with
pEJ-ras. EL-4/E7 is the thymoma EL-4 retrovirally transduced with E7.
47

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
L. monocytogenes strains and propagation
1001881Listeria strains used were Lm-LLO-E7 (hly-E7 fusion gene in an episomal
expression
system; Figure 1A), Lm-E7 (single-copy E7 gene cassette integrated into
Listeria genome),
Lm-LLO-NP ("DP-L2028"; hly-NP fusion gene in an episomal expression system),
and Lm-
Gag ("ZY-18"; single-copy HIV-1 Gag gene cassette integrated into the
chromosome). E7
was amplified by PCR using the primers 5'-GGCTCGAGCATGGAGATACACC-3 (SEQ ID
No: 17; XhoI site is underlined) and 5'-GGGGACTAGTTTATGGTTTCTGAGAACA-3'
(SEQ ID No: 18; SpeI site is underlined) and ligated into pCR2.1 (Invitrogen,
San Diego,
CA). E7 was excised from pCR2.1 by XhoI/ SpeI digestion and ligated into pGG-
55. The hly-
E7 fusion gene and the pluripotential transcription factor PrfA were cloned
into pAM401, a
multicopy shuttle plasmid (Wirth R et al, J Bacteriol, 165: 831, 1986),
generating pGG-55.
The hly promoter drives the expression of the first 441 AA of the hly gene
product, (lacking
the hemolytic C-terminus, referred to below as "ALLO," and having the sequence
set forth in
SEQ ID No: 25), which is joined by the XhoI site to the E7 gene, yielding a
hly-E7 fusion
gene that is transcribed and secreted as LLO-E7. Transformation of a PrfA
negative strain of
Listeria, XFL-7 (provided by Dr. Hao Shen, University of Pennsylvania), with
pGG-55
selected for the retention of the plasmid in vivo (Figures 1A-B). The hly
promoter and gene
fragment were generated using primers 5'-
GGGGGCTAGCCCTCCTTTGATTAGTATATTC-3' (SEQ ID No: 19; NheI site is
underlined) and 5'-CTCCCTCGAGATCATAATTTACTTCATC-3' (SEQ ID No: 20; XhoI
site is underlined). The PifA gene was PCR amplified using primers 5'-
GACTACAAGGACGATGACCGACAAGTGATAACCCGGGATCTAAATAAATCCGTT
T-3' (SEQ ID No: 27; XbaI site is underlined)
and 5'-
CCCGTCGACCAGCTCTTCTTGGTGAAG-3' (SEQ ID No: 21; Sall site is underlined). Lm-
E7 was generated by introducing an expression cassette containing the hly
promoter and
signal sequence driving the expression and secretion of E7 into the orfZ
domain of the LM
genome. E7 was amplified by PCR using the primers 5'-
GCGGATCCCATGGAGATACACCTAC-3' (SEQ ID No: 22; BamHI site is underlined) and
5'-GCTCTAGATTATGGTTTCTGAG-3' (SEQ ID No: 23; XbaI site is underlined). E7 was
then ligated into the pZY-21 shuttle vector. LM strain 10403S was transformed
with the
resulting plasmid, pZY-21-E7, which includes an expression cassette inserted
in the middle of
a 1.6-kb sequence that corresponds to the orfX, Y, Z domain of the LM genome.
The
homology domain allows for insertion of the E7 gene cassette into the orfZ
domain by
homologous recombination. Clones were screened for integration of the E7 gene
cassette into
48

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
the orfZ domain. Bacteria were grown in brain heart infusion medium with (Lm-
LLO-E7 and
Lm-LLO-NP) or without (Lm-E7 and ZY-18) chloramphenicol (20 pg/ml). Bacteria
were
frozen in aliquots at -80 C. Expression was verified by Western blotting
(Figure 2).
Western blotting
[001891Listeria strains were grown in Luria-Bertoni medium at 37 C and were
harvested at
the same optical density measured at 600 nm. The supernatants were TCA
precipitated and
resuspended in lx sample buffer supplemented with 0.1 N NaOH. Identical
amounts of each
cell pellet or each TCA-precipitated supernatant were loaded on 4-20% Tris-
glycine SDS-
PAGE gels (NO VEX, San Diego, CA). The gels were transferred to polyvinylidene
difluoride
and probed with an anti-E7 monoclonal antibody (mAb) (Zymed Laboratories,
South San
Francisco, CA), then incubated with HRP-conjugated anti-mouse secondary Ab
(Amersham
Pharmacia Biotech, Little Chalfont, U.K.), developed with Amersham ECL
detection
reagents, and exposed to Hyperfilm (Amersham Pharmacia Biotech).
Measurement of tumor growth
[001901Tumors were measured every other day with calipers spanning the
shortest and
longest surface diameters. The mean of these two measurements was plotted as
the mean
tumor diameter in millimeters against various time points. Mice were
sacrificed when the
tumor diameter reached 20 mm. Tumor measurements for each time point are shown
only for
surviving mice.
Effects of Listeria recombinants on established tumor growth
[001911Six- to 8-wk-old C57BL/6 mice (Charles River) received 2 x 105 TC-1
cells s.c. on
the left flank. One week following tumor inoculation, the tumors had reached a
palpable size
of 4-5 mm in diameter. Groups of eight mice were then treated with 0.1 LD50
i.p. Lm-LLO-
E7 (107 CFU), Lm- E7 (106 CFU), Lm-LLO-NP (107 CFU), or Lm-Gag (5 x 105 CFU)
on
days 7 and 14.
51Cr release assay
11001921057BL/6 mice, 6-8 wk old, were immunized i.p. with 0.1LD50 Lm-LLO-E7,
Lm-E7,
Lm-LLO-NP, or Lm-Gag. Ten days post-immunization, spleens were harvested.
Splenocytes
were established in culture with irradiated TC-1 cells (100:1, splenocytes:TC-
1) as feeder
cells; stimulated in vitro for 5 days, then used in a standard 51Cr release
assay, using the
following targets: EL-4, EL-4/E7, or EL-4 pulsed with E7 H-2b peptide
(RAHYNIVTF). E:T
cell ratios, performed in triplicate, were 80:1, 40:1, 20:1, 10:1, 5:1, and
2.5:1. Following a 4-h
incubation at 37 C, cells were pelleted, and 50 p 1 supernatant was removed
from each well.
Samples were assayed with a Wallac 1450 scintillation counter (Gaithersburg,
MD). The
49

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
percent specific lysis was determined as [(experimental counts per minute
(cpm)- spontaneous
cpm)/(total cpm - spontaneous cpm)] x 100.
TC-1-specific proliferation
[00193] C57BL/6 mice were immunized with 0.1 LD50 and boosted by i.p.
injection 20 days
later with 1 LD50 Lm-LLO-E7, Lm-E7, Lm-LLO-NP, or Lm-Gag. Six days after
boosting,
spleens were harvested from immunized and naive mice. Splenocytes were
established in
culture at 5 x 105/well in flat-bottom 96-well plates with 2.5 x 104, 1.25 x
104, 6 x 103, or 3 x
103 irradiated TC-1 cells/well as a source of E7 Ag, or without TC-1 cells or
with 10 p g/ml
Con A. Cells were pulsed 45 h later with 0.5 p Ci [3H]thymidine/well. Plates
were harvested
18 h later using a Tomtec harvester 96 (Orange, CT), and proliferation was
assessed with a
Wallac 1450 scintillation counter. The change in cpm was calculated as
experimental cpm -
no Ag cpm.
Flow cytometric analysis
[001941057BL/6 mice were immunized intravenously (i.v.) with 0.1 LD50 Lm-LLO-
E7 or
Lm-E7 and boosted 30 days later. Three-color flow cytometry for CD8 (53-6.7,
PE
conjugated), CD62 ligand (CD62L; MEL-14, APC conjugated), and E7 H-2Db
tetramer was
performed using a FACSCalibur flow cytometer with CellQuest software (Becton

Dickinson, Mountain View, CA). Splenocytes harvested 5 days after the boost
were stained at
room temperature (rt) with H-2Db tetramers loaded with the E7 peptide
(RAHYNIVTF) or a
control (HIV-Gag) peptide. Tetramers were used at a 1/200 dilution and were
provided by Dr.
Larry R. Pease (Mayo Clinic, Rochester, MN) and by the NIAID Tetramer Core
Facility and
the NIH AIDS Research and Reference Reagent Program. Tetramer4, CD84, CD62L10w
cells
were analyzed.
B16FO-Ova experiment
1100195124 C57BL/6 mice were inoculated with 5 x 105 B16FO-Ova cells. On days
3, 10 and
17, groups of 8 mice were immunized with 0.1 LD50 Lm-OVA (106 cfu), Lm-LLO-OVA
(108
cfu) and eight animals were left untreated.
Statistics
11001961 For comparisons of tumor diameters, mean and SD of tumor size for
each group were
determined, and statistical significance was determined by Student's t test. p
< 0.05 was
considered significant.
RESULTS
[00197]Lm-E7 and Lm-LLO-E7 were compared for their abilities to impact on TC-1
growth.
Subcutaneous tumors were established on the left flank of C57BL/6 mice. Seven
days later

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
tumors had reached a palpable size (4-5 mm). Mice were vaccinated on days 7
and 14 with
0.1 LD50 Lm-E7, Lm-LLO-E7, or, as controls, Lm-Gag and Lm-LLO-NP. Lm-LLO-E7
induced complete regression of 75% of established TC-1 tumors, while tumor
growth was
controlled in the other 2 mice in the group (Figure 3). By contrast,
immunization with Lm-E7
and Lm-Gag did not induce tumor regression. This experiment was repeated
multiple times,
always with very similar results. In addition, similar results were achieved
for Lm-LLO-E7
under different immunization protocols. In another experiment, a single
immunization was
able to cure mice of established 5 mm TC-1 tumors.
1001981In other experiments, similar results were obtained with 2 other E7-
expres sing tumor
cell lines: C3 and EL-4/E7. To confirm the efficacy of vaccination with Lm-LLO-
E7, animals
that had eliminated their tumors were re-challenged with TC-1 or EL-4/E7 tumor
cells on day
60 or day 40, respectively. Animals immunized with Lm-LLO-E7 remained tumor
free until
termination of the experiment (day 124 in the case of TC-1 and day 54 for EL-
4/E7).
1001991Thus, expression of an antigen as a fusion protein with ALLO enhances
the
immunogenic ity of the antigen..
EXAMPLE 2: LM-LLO-E7 TREATMENT ELICITS TC-1 SPECIFIC
SPLENOCYTE PROLIFERATION
1002001To measure induction of T cells by Lm-E7 with Lm-LLO-E7, TC-1-specific
proliferative responses, a measure of antigen-specific immunocompetence, were
measured in
immunized mice. Splenocytes from Lm-LLO-E7-immunized mice proliferated when
exposed
to irradiated TC-1 cells as a source of E7, at splenocyte: TC-1 ratios of
20:1, 40:1, 80:1, and
160:1 (Figure 4). Conversely, splenocytes from Lm-E7 and rLm control-immunized
mice
exhibited only background levels of proliferation.
EXAMPLE 3: FUSION OF E7 TO LLO, ActA, OR A PEST AMINO ACID
SEQUENCE ENHANCES E7-SPECIFIC IMMUNITY AND GENERATES TUMOR-
INFILTRATING E7-SPECIFIC CD8+ CELLS
MATERIALS AND EXPERIMENTAL METHODS
1002011500 mcl (microliter) of MATRIGEL , comprising 100 mcl of 2 x 105 TC-1
tumor
cells in phosphate buffered saline (PBS) plus 400 mcl of MATRIGEL (BD
Biosciences,
Franklin Lakes, N.J.) were implanted subcutaneously on the left flank of 12
C57BL/6 mice
(n=3). Mice were immunized intraperitoneally on day 7, 14 and 21, and spleens
and tumors
51

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
were harvested on day 28. Tumor MATRIGELs were removed from the mice and
incubated
at 4 C overnight in tubes containing 2 milliliters (ml) of RP 10 medium on
ice. Tumors were
minced with forceps, cut into 2 mm blocks, and incubated at 37 C for 1 hour
with 3 ml of
enzyme mixture (0.2 mg/ml collagenase-P, 1 mg/ml DNAse-1 in PBS). The tissue
suspension
was filtered through nylon mesh and washed with 5% fetal bovine serum + 0.05%
of NaN3 in
PBS for tetramer and IFN-gamma staining.
[00202] Splenocytes and tumor cells were incubated with 1 micromole (mcm) E7
peptide for 5
hours in the presence of brefeldin A at 107 cells/ml. Cells were washed twice
and incubated in
50 mcl of anti-mouse Fc receptor supernatant (2.4 G2) for 1 hour or overnight
at 4 C. Cells
were stained for surface molecules CD8 and CD62L, permeabilized, fixed using
the
permeabilization kit Golgi-stop or Golgi-Plug (Pharmingen, San Diego,
Calif.), and
stained for IFN-gamma. 500,000 events were acquired using two-laser flow
cytometer
FACSCalibur and analyzed using Cellquest Software (Becton Dickinson, Franklin
Lakes,
NJ). Percentages of IFN-gamma secreting cells within the activated (CD62L10v)
CD8 + T cells
were calculated.
11002031 For tetramer staining, H-2D6 tetramer was loaded with phycoerythrin
(PE)-conjugated
E7 peptide (RAHYNIVTF, SEQ ID NO: 24), stained at A for 1 hour, and stained
with anti-
allophycocyanin (APC) conjugated MEL-14 (CD62L) and FITC-conjugated CD8 at 4
C
for 30 mm. Cells were analyzed comparing tetramer+CD8+ CD62L1' cells in the
spleen and
in the tumor.
RESULTS
[00204] To analyze the ability of Lm-ActA-E7 to enhance antigen specific
immunity, mice
were implanted with TC-1 tumor cells and immunized with either Lm-LLO-E7 (1 x
107
CFU), Lm-E7 (1 x 106 CFU), or Lm-ActA-E7 (2 x 108 CFU), or were untreated
(naïve).
Tumors of mice from the Lm-LLO-E7 and Lm-ActA-E7 groups contained a higher
percentage of IFN-gamma-secreting CD8 + T cells (Figure 5A) and tetramer-
specific CD8+
cells (Figure 5B) than in Lm-E7 or naive mice.
11002051 In another experiment, tumor-bearing mice were administered Lm-LLO-
E7, Lm-
PEST-E7, Lm-APEST-E7, or Lm-E7epi, and levels of E7-specific lymphocytes
within the
tumor were measured. Mice were treated on days 7 and 14 with 0.1 LD50 of the 4
vaccines.
Tumors were harvested on day 21 and stained with antibodies to CD62L, CD8, and
with the
E7/Db tetramer. An increased percentage of tetramer-positive lymphocytes
within the tumor
52

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
were seen in mice vaccinated with Lm-LLO-E7 and Lm-PEST-E7 (Figure 6A). This
result
was reproducible over three experiments (Figure 6B).
[00206] Thus, Lm-LLO-E7, Lm-ActA-E7, and Lm-PEST-E7 are each efficacious at
induction
of tumor-infiltrating CD8+ T cells and tumor regression.
EXAMPLE 4: PASSAGING OF LISTERIA VACCINE VECTORS THROUGH MICE
ELICITS INCREASED IMMUNE RESPONSES TO HETEROLOGOUS AND
ENDOGENOUS ANTIGENS
MATERIALS AND EXPERIMENTAL METHODS
Bacterial Strains
[00207]L. monocytogenes strain 10403S, serotype 1 (ATCC, Manassas, Va.) was
the wild
type organism used in these studies and the parental strain of the constructs
described below.
Strain 10403S has an LD50 of approximately 5 x 104 CFU when injected
intraperitoneally into
BALB/c mice. "Lm-Gag" is a recombinant LM strain containing a copy of the HIV-
1 strain
HXB (subtype B laboratory strain with a syncytia-forming phenotype) gag gene
stably
integrated into the listerial chromosome using a modified shuttle vector
pKSV7. Gag protein
was expressed and secreted by the strain, as determined by Western blot. All
strains were
grown in brain-heart infusion (BHI) broth or agar plates (Difco Labs, Detroit,
Mich).
Bacterial Culture
[00208]Bacteria from a single clone expressing the passenger antigen and/or
fusion protein
were selected and cultured in BHI broth overnight. Aliquots of this culture
were frozen at -
70 C with no additives. From this stock, cultures were grown to 0.1-0.2 O.D.
at 600 nm, and
aliquots were again frozen at -70 C with no additives. To prepare cloned
bacterial pools, the
above procedure was used, but after each passage a number of bacterial clones
were selected
and checked for expression of the target antigen, as described herein. Clones
in which
expression of the foreign antigen was confirmed were used for the next
passage.
Passage of Bacteria in Mice
[00209[6-8 week old female BALB/c (H-2d) mice were purchased from Jackson
Laboratories
(Bar Harbor, Me) and were maintained in a pathogen-free microisolator
environment. The
titer of viable bacteria in an aliquot of stock culture, stored frozen at -70
C, was determined
by plating on BHI agar plates on thawing and prior to use. In all, 5 x 105
bacteria were
injected intravenously into BALB/c mice. After 3 days, spleens were harvested,

homogenized, and serial dilutions of the spleen homogenate were incubated in
BHI broth
53

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
overnight and plated on BHI agar plates. For further passage, aliquots were
again grown to
0.1-0.2 0.D., frozen at -70 C, and bacterial titer was again determined by
serial dilution.
After the initial passage (passage 0), this sequence was repeated for a total
of 4 times.
Intracellular Cytokine Stain for IFN-Gamma
1002101Lymphocytes were cultured for 5 hours in complete RPMI-10 medium
supplemented
with 50 U/ml human recombinant IL-2 and 1 microliter/ml Brefeldin A
(GolgistopTm;
PharMingen, San Diego, CA) in the presence or absence of either the cytotoxic
T-cell (CTL)
epitope for HIV-GAG (AMQMLKETI; SEQ ID No: 25), Listeria LLO (GYKDGNEYI; SEQ
ID No: 26) or the HPV virus gene E7 (RAHYNIVTF (SEQ ID No: 24), at a
concentration of
1 micromole. Cells were first surface-stained, then washed and subjected to
intracellular
cytokine stain using the Cytofix/Cytoperm kit in accordance with the
manufacturer's
recommendations (PharMingen, San Diego, CA). For intracellular IFN-gamma
stain, FITC-
conjugated rat anti-mouse IFN-gamma monoclonal antibody (clone XMG 1.2) and
its isotype
control Ab (rat IgGl; both from PharMingen) was used. In all, 106 cells were
stained in PBS
containing 1% Bovine Serum Albumin and 0.02% sodium azide (FACS Buffer) for 30
minutes at 4 C. followed by 3 washes in FACS buffer. Sample data were
acquired on either a
FACScanTm flowcytometer or FACSCaliburTm instrument (Becton Dickinson, San
Jose, CA).
Three-color flow cytometry for CD8 (PERCP conjugated, rat anti-mouse, clone 53-
6.7
Pharmingen, San Diego, Calif.), CD62L (APC conjugated, rat anti-mouse, clone
MEL-14),
and intracellular IFN-gamma was performed using a FACSCaliburTm flow
cytometer, and
data were further analyzed with CELLQuest software (Becton Dickinson, Mountain
View,
CA). Cells were gated on CD8 high and CD62L1' before they were analyzed for
CD8 + and
intracellular IFN-gamma staining.
RESULTS
Passaging in Mice Increases the Virulence of Recombinant Listeria
Monocvtogenes
1002111Three different constructs were used to determine the impact of
passaging on
recombinant Listeria vaccine vectors. Two of these constructs carry a genomic
insertion of
the passenger antigen: the first comprises the HIV gag gene (Lm-Gag), and the
second
comprises the HPV E7 gene (Lm-E7). The third (Lm-LLO-E7) comprises a plasmid
with the
fusion gene for the passenger antigen (HPV E7) fused with a truncated version
of LLO and a
gene encoding PrfA, the positive regulatory factor that controls Listeria
virulence factors.
This plasmid was used to complement a PrfA negative mutant so that in a live
host, selection
pressures would favor conservation of the plasmid, because without it the
bacterium is
54

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
avirulent. All 3 constructs had been propagated extensively in vitro for many
bacterial
generations.
1002121Passaging the bacteria resulted in an increase in bacterial virulence,
as measured by
numbers of surviving bacteria in the spleen, with each of the first 2
passages. For Lm-Gag and
Lm-LLO-E7, virulence increased with each passage up to passage 2 (Figure 7A).
The
plasmid-containing construct, Lm-LLO-E7, demonstrated the most dramatic
increase in
virulence. Prior to passage, the initial immunizing dose of Lm-LLO-E7 had to
be increased to
107 bacteria and the spleen had to be harvested on day 2 in order to recover
bacteria (whereas
an initial dose of 105 bacteria for Lm-Gag was harvested on day 3). After the
initial passage,
the standard dosage of Lm-LLO-E7 was sufficient to allow harvesting on day 3.
For Lm-E7,
virulence increased by 1.5 orders of magnitude over unpassaged bacteria
(Figure 7B).
1002131Thus, passage through mice increases the virulence of Listeria vaccine
strains.
Passa2in2 Increases the Ability of L. monocyto2enes to Induce CD8+ T Cells
1002141Next, the effect of passaging on induction of antigen-specific CD8+ T
cells was
determined by intracellular cytokine staining with immunodominant peptides
specific for
MHC-class I using HIV-Gag peptide AMQMLKETI (SEQ ID No: 25) and LLO 91-99
(GYKDGNEYI; SEQ ID No: 26). Injection of 103 CFU passaged bacteria (Lm-Gag)
into
mice elicited significant numbers of HIV-Gag-specific CD8+ T cells, while the
same dose of
non-passaged Lm-Gag induced no detectable Gag-specific CD8+ T cells. Even
increasing the
dose of unpassaged bacteria 100-fold did not compensate for their relative
avirulence; in fact,
no detectable Gag-specific CD8+ T cells were elicited even at the higher dose.
The same dose
increase with passaged bacteria increased Gag-specific T cell induction by 50%
(Figure 8).
The same pattern of induction of antigen-specific CD8+ T cells was observed
with LLO-
specific CD8+ T cells, showing that these results were not caused by the
properties of the
passenger antigen, since they were observed with LLO, an endogenous Listeria
antigen.
1002151Thus, passage through mice increases the immunogenicity of Listeria
vaccine strains.
EXAMPLE 5: A PrfA-CONTAINING PLASMID IS STABLE IN AN LM STRAIN
WITH A PrfA DELETION IN THE ABSENCE OF ANTIBIOTICS
MATERIALS AND EXPERIMENTAL METHODS
Bacteria
1002161L. monocytogenes strain XFL7 contains a 300 base pair deletion in the
PrfA gene
XFL7 carries pGG55 which partially restores virulence and confers CAP
resistance, and is
described in United States Patent Application Publication No. 200500118184.

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
Development of protocol for plasmid extraction from Listeria
[00217[1 mL of Listeria monocytogenes Lm-LLO-E7 research working cell bank
vial was
inoculated into 27 mL BH1 medium containing 34 p g/mL CAP and grown for 24
hours at
37 C and 200 rpm.
[00218] Seven 2.5 mL samples of the culture were pelleted (15000 rpm for 5
minutes), and
pellets were incubated at 37 C with 50 p 1 lysozyme solution for varying
amounts of time,
from 0-60 minutes.
[00219] Lysozyme solution:
- 29 pl 1 M dibasic Potassium Phosphate
- 21 pl 1 M monobasic Potassium Phosphate
- 500 pl 40% Sucrose (filter sterilized through 0.45 /p m filter)
- 450 pl water
- 60 pl lysozyme (50 mg/mL)
[00220] After incubation with the lysozyme, the suspensions were centrifuged
as before and
the supernatants discarded. Each pellet was then subjected to plasmid
extraction by a
modified version of the QIAprep Spin Miniprep Kit (Qiagen, Germantown,
Maryland)
protocol. The changes to the protocol were as follows:
1. The volumes of buffers PI, P2 and N3 were all increased threefold to allow
complete lysis of the increased biomass.
2. 2 mg/mL of lysozyme was added to the resuspended cells before the addition
of P2. The lysis solution was then incubated at 37 C for 15 minutes before
neutralization.
3. The
plasmid DNA was resuspended in 30 p L rather than 50 p L to increase the
concentration.
11002211 In other experiments, the cells were incubated for 15min in P1 buffer
+ Lysozyme,
then incubated with P2 (lysis buffer) and P3 (neutraliztion buffer) at room
temperature.
[00222] Equal volumes of the isolated plasmid DNA from each subculture were
run on an
0.8% agarose gel stained with ethidium bromide and visualized for any signs of
structural or
segregation instability.
[00223] The results showed that plasmid extraction from L. monocytogenes Lm-
LLO-E7
increases in efficiency with increasing incubation time with lysozyme, up to
an optimum level
at approximately 50 minutes incubation.
[00224] These results provide an effective method for plasmid extraction from
Listeria
vaccine strains.
56

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
Replica plating
11002251 Dilutions of the original culture were plated onto plates containing
LB or TB agar in
the absence or presence of 34 p g/mL CAP. The differences between the counts
on selective
and non-selective agar were used to determine whether there was any gross
segregational
instability of the plasmid.
RESULTS
[00226] The genetic stability (i.e. the extent to which the plasmid is
retained by or remains
stably associated with the bacteria in the absence of selection pressure; e.g.
antibiotic
selection pressure) of the pGG55 plasmid in L. monocyto genes strain XFL7 in
the absence of
antibiotic was assessed by serial sub-culture in both Luria-Bertani media (LB:
5 g/L NaC1, 10
g/ml soy peptone, 5 g/L yeast extract) and Terrific Broth media (TB: 10 g/L
glucose, 11.8 g/L
soy peptone, 23.6 g/L yeast extract, 2.2 g/L KH2PO4, 9.4 g/L K2HPO4), in
duplicate cultures.
50 mL of fresh media in a 250 mL baffled shake flask was inoculated with a
fixed number of
cells (1 0DmL), which was then subcultured at 24 hour intervals. Cultures were
incubated in
an orbital shaker at 37 C and 200 rpm. At each subculture the 0D600 was
measured and used
to calculate the cell doubling time (or generation) elapsed, until 30
generations were reached
in LB and 42 in TB. A known number of cells (15 0DmL) at each subculture stage

(approximately every 4 generations) were pelleted by centrifugation, and the
plasmid DNA
was extracted using the Qiagen QIAprep Spin Miniprep protocol described
above. After
purification, plasmid DNA was subjected to agarose gel electrophoresis,
followed by
ethidium bromide staining. While the amount of plasmid in the preps varied
slightly between
samples, the overall trend was a constant amount of plasmid with respect to
the generational
number of the bacteria (Figures 9A-B). Thus, pGG55 exhibited stability in
strain XFL7, even
in the absence of antibiotic.
[00227]Plasmid stability was also monitored during the stability study by
replica plating on
agar plates at each stage of the subculture. Consistent with the results from
the agarose gel
electrophoresis, there was no overall change in the number of plasmid-
containing cells
throughout the study in either LB or TB liquid culture (Figures 10 and 11,
respectively).
[00228] These findings demonstrate that PrfA-encoding plasmids exhibit
stability in the
absence of antibiotic in Listeria strains containing mutations in d'A gene.
MATERIALS AND METHODS (examples 6-10)
[00229]PCR reagents:
[00230] The primers used for amplification of the PrfA gene and discrimination
of the Dl 33V
57

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
mutation are shown in Table 1. Stock solutions of the primers ADV451, 452 and
453 were
prepared by diluting the primers in TE buffer to 400 M. An aliquot of the
stock solution was
further diluted to 20 M in water (PCR grade) to prepare a working solution.
Primers were
stored at -20 C. The reagents used in the PCR are shown in Table 2.
[00231] Table 1. Primers ADV451, 452 and 453.
Primer Orientation Sequence (5' ¨> 3') Specificity
ADV451 Forward CCTAGCTAAATTTAATGT D133V mutation
(SEQ ID NO: 28)
ADV452 Forward CCTAGCTAAATTTAATGA Wild-type sequence
(SEQ ID NO: 29)
ADV453 Reverse TAATTTTCCCCAAGTAGCAGG Shared sequence
(SEQ ID NO: 30)
[00232] Table 2. PCR reagents.
Description Provider Catalog number
1 0.2 ml thin-walled PCR tubes: GeneAmp Applied N801-0612
autoclaved reaction tube with cap Biosystems
2 Water (PCR reagent) Sigma W1754
3 Taq DNA Polymerase with 10x reaction buffer Sigma D1806
containing 15 mM MgC12
4 Set of deoxynucleotides (dNTPs), 10 mM each Sigma D7295
5 Primers ADV451, ADV452 and ADV453 Invitrogen
6 Template DNA, midipreparations of pGG55
plasmids
7 Thermal cycler PTC200 (48 wells block) MJ Research
Plasmid DNA preparation
[002331pGG55 plasmids with (pGG55 D133V) and without (pGG55 WT) the prfA
mutation
were extracted and purified by midipreparations either from E. coli or
Listeria monocyto genes
using the PureLinkTM HiPure Plasmid Midiprep Kit (Invitrogen, K2100-05),
according to the
manufacturer's instructions. For plasmid purification from Listeria, bacterial
strains carrying
the pGG55 D133V or WT plasmids were streak plated from frozen stocks in BHI
agar plates
supplemented with chloramphenicol (25 g/m1). A single colony from each strain
was grown
in 5 ml of selective medium (BHI broth with 25 g/m1 of chloramphenicol) for 6
hours with
vigorous shaking at 37 C and subinoculated 1:500 in 100 ml of selective medium
for
overnight growth under similar conditions. Bacteria from the overnight culture
were harvested
by centrifugation at 4,000 x g for 10 minutes and resuspended buffer R3
(resuspension buffer)
58

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
containing 2 mg/ml of lysozyme (Sigma, L7001). The bacteria suspension was
incubated for
at least 1 hour at 37 C before proceeding to the regular protocol.
Concentration and purity of
the eluted plasmids were measured in a spectrophotometer at 260nm and 280nm.
To prepare
the template DNAs, the pGG55 D133V and WT plasmids were resuspended in water
to a
final concentration of 1 ng/n1 from the midiprep stock solution. For the pGG55
WT plasmid,
serial 10-fold dilutions from the 1 ng/n1 solution were prepared,
corresponding to dilutions
from 104 to 10-7.
prfA specific PCR protocol to test clinical grade material
[00234]The reaction mixture contained lx PCR buffer, 1.5 mM MgC12, 0.8 mM
dNTPs, 0.4
M of each primer, 0.05 U/n1 of Taq DNA polymerase and 0.04 ng/n1 of the pGG55
D133V
template plasmid. For each test, 10 tubes were required and the key components
in each tube
in a 25 n1 reaction are shown in the Table 3. For the PCR reaction, a master
mix was prepared
with enough reagents for 11 reactions as shown in Table 4, and 24 n1 of this
PCR mix was
added to each tube. Subsequently, a total of 1 n1 of the serially diluted
pGG55 WT plasmid
was added to the corresponding tubes: 1 ng in tube 3; 100 pg in tube 4; 10 pg
in tube 5; 1 pg
in tube 6; 100 fg in tube 7; 10 fg in tube 8; 1 fg in tube 9; 0.1 fg in tube
10. This serial dilution
was used to calibrate a standard curve to determine the method sensitivity.
Additionally, 0.5
n1 of water and 0.5 n1 of primer ADV451 (20 M stock) were added in tube 1,
and 1 n1 of
water added in tube 2, completing 25 n1 of final volume. The quantities of
each reagent per
tube for a 25 n1 reaction are shown in Table 5. The PCR cycling conditions
used in the
reaction are shown in Table 6.
[00235]After conclusion of the PCR reaction, 5 n1 of gel-loading buffer (6x,
with
bromophenol blue) was added to each sample and 10 n1 were analyzed by
electrophoresis in
1.2% agarose gel in TBE buffer. The gel dimensions were 7 cm x 7 cm x 1 cm
with a 15
sample wells (1 mm x 2 mm) comb. The gel was run at 100 V for ¨30 minutes,
until the
bromophenol blue dye reached the middle of the gel. The gel was stained in
ethidium bromide
(0.5 OM) for 20 minutes, destaining in water for 10 minutes. The gel is
visualized by
illumination with UV light and photographed. The image was analyzed using a
band
densitometry software (Quantity One version 4.5.1, BioRad).
[00236] Table 3. Set of individual PCR reactions to validate the method to
detect the presence
of wild-type prfA sequence in Lm-LLO-E7 samples.
Tube Primer A Primer B Template DNA Function Expected
result
1 ADV451 ADV453 1 ng of p0055 Positive control for Positive
(D133V) the ADV451 reaction
59

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
2 ADV452 ADV453 1 ng of p0055 Negative control for Negative
(D133V) the ADV452 reaction
(specificity)
3 ADV452 ADV453 1 ng of p0055 Positive control for Positive
(wild-type) + 1 ng the ADV452 reaction
of p0055 (D133V)
4 ADV452 ADV453 100 pg of p0055 Test the sensitivity of Positive
(wild-type) +1 ng the reaction
of p0055 (D133V)
ADV452 ADV453 10 pg of p0055 Test the sensitivity of Positive
(wild-type) + 1 ng the reaction
of p0055 (D133V)
6 ADV452 ADV453 1 pg of p0055 Test the sensitivity of Positive
(wild-type) + 1 ng the reaction
of p0055 (D133V)
7 ADV452 ADV453 100 fg of p0055 Test the sensitivity of Positive
(wild-type) + lng the reaction
p0055 (D133V)
8 ADV452 ADV453 10 fg of p0055 Test the sensitivity of Positive
(wild-type) + the reaction
p0055 (D133V)
9 ADV452 ADV453 1 fg of p0055 Test the sensitivity of Weakly
(wild-type) + the reaction positive
p0055 (D133V)
ADV452 ADV453 0.1 fg of p0055 Test the sensitivity of To be
(wild-type) + the reaction determined
p0055 (D133V)
[00237] Table 4. Master PCR mix preparation.
Reagent Quantity ( 1)
Water 206.25
Taq DNA Polymerase 10x reaction buffer 27.5
containing 15 mM MgC12
Deoxynucleotides (dNTPs) 10 mM each 5.5
Primers ADV452 (20 M in water) 5.5
Primers ADV453 (20 M in water) 5.5
pGG55 D133V (Lm-LLO-E7) plasmid (1 ng/n1) 11
Taq DNA Polymerase (5 U/ 1) 2.75
Total 264
11002381 Table 5. PCR protocol for validation of the method to detect the
presence of wild-
5 type pifA sequence using primers ADV451, 452 and 453.
Reagent PCR
Water 18.75 n1
PCR Buffer 10x + MgC12 15mM 2.5 n1
Deoxynucleotides mix (dATP, dCTP, dGTP and dTTP) 0.5 n1
10mM each
Primer ADV452 (20 M) 0.5 n1
Primer ADV453 (20 M) 0.5 n1
Taq DNA polymerase (5 U/n1) 0.25 n1

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
Template DNA (1 ng/n1) pGG55 D133V 1 n1
Template DNA pGG55 WT (tubes 3 to 10)a 1 IA
Final volume per tubeb 25 n1
a pGG55 WT (1 ng in tube 3; 100 pg in tube 4; 10 pg in tube 5; 1 pg in tube 6;
100 fg in tube 7; 10
fg in tube 8; 1 fg in tube 9; 0.1 fg in tube 10).
b In tube 1, add 0.5 pi of water and 0.5 pi of primer ADV451 (20 1..iM stock);
in tube 2 add 1 1.11 of
water.
[00239] Table 6. PCR cycling conditions to detect the presence of wild-type
prfA sequence
using primers ADV451, 452 and 453.
Step Temperature Time Number of cycles
1. 94 C 2 minutes and 30
seconds 1
2. 94 C 30 seconds 1
3. 53 C 30 seconds 1
4. 72 C 30 seconds 1
5. Repeat steps 2 to 4 12
6. 94 C 30 seconds 1
7. 50 C 30 seconds 1
8. 72 C 30 seconds 1
9. Repeat steps 6 to 8 23
10. 72 C 10 minutes 1
Sequencing:
[00240] Sequencing of the plasmids was done using the dideoxy sequencing
method. The
plasmids pGG55 D133V and pGG55 WT were mixed at different ratios (1:1, 1:10,
1;100,
1:1,000 and 1:10,000). The total amount of plasmid in the mixture was kept
constant (500 rig)
and the plasmid containing the wild-type sequence was 10-fold serially diluted
in relation to
the D133V plasmid to determine the sensitivity of the method.
RESULTS
EXAMPLE 6: SEQUENCING IS NOT A SENSITIVE METHOD TO DETECT THE
REVERSION OF THE D133V MUTATION.
[00241] To estimate the sensitivity of sequencing in detecting the wild-type
prfA sequence, the
pGG55 D133V and WT plasmids were mixed at the different ratios and sequenced.
The
results are shown in Figure 12 and reveal that sequencing has a high
specificity in
discriminating the prfA D133V mutation (Figure 12). On the other hand, the
sensitivity is low
and the maximum dilution of wild-type prfA pGG55 plasmid with a detectable
peak in the
sequence was 1 in 10 (Figure 12). In conclusion, although sequencing is very
specific, the
sensitivity of the method is low and not appropriate to screen for the
presence of rare events
61

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
such as revertants of the prfA D133V mutation in Lm-LLO-E7 samples.
EXAMPLE 7: DEVELOPMENT OF A HIGHLY SPECIFIC AND SENSITIVE PCR
METHOD TO DETECT REVERSION OF THE D133V MUTATION.
[00242] Given the low sensitivity of sequencing to detect rare events, it
became imperative to
develop a more sensitive method with similar specificity to detect reversion
of the D133V
mutation to wild-type. To achieve this goal, we designed a PCR-based method
that
specifically amplifies the wild-type sequence and is sensitive enough to
detect at least 1 wild-
type copy of pifA in 10,000,000 copies of the D133V mutated sequence. We
designed 3
primers for this method: ADV451, ADV452 and ADV453 (Table 1). Both ADV451 and
ADV452 are forward primers and differ in the last nucleotide at the 3'
position to
discriminate the A¨>T (D133V) mutation at position 398 of the prfA gene. The
ADV453
primer is the reverse primer located approximately 300 bp downstream the
annealing site of
the ADV451 and ADV452 primers (Figure 13). The expected PCR band obtained with
the
primers ADV451 or ADV452 and ADV453 is 326 bp. Under stringent conditions, the
ADV451 primer should only amplify the pGG55 D133V plasmid, whereas the ADV452
would be specific to the wild-type prfA sequence.
EXAMPLE 8: SPECIFICITY OF THE PCR METHOD.
[00243] The reaction using the primer ADV451 was very specific and amplified
the mutated
D133V prfA sequence (lanes 1 to 3), but not the wild-type sequence (lanes 4 to
6). However, a
very faint band can be detected in lane 4, when 5 ng of template DNA was used,
but not with
1 ng (Figure 14).
[00244] As shown in Figure 15, the reaction with the ADV452 primer only
amplified the wild-
type prfA sequence (lanes 4, 5 and 6), and no bands were detected when the
pGG55 carrying
the D133V prfA mutation was used as a template (lanes 1, 2 and 3), even when
using 5 ng of
plasmid in the reaction (Figure 16). In conclusion, the PCR reactions with
primers ADV451
and ADV452 are very specific and able to discriminate the A->T (D133V)
mutation at
position 398 of the prfA gene in the pGG55 plasmid. Based on these results, we
selected the
amount of 1 ng as the standard amount of template DNA to be used in the
reaction.
EXAMPLE 9: SENSITIVITY OF THE PCR METHOD.
[00245]The sensitivity of the reaction was tested using 1 ng of template DNA.
For the
plasmid carrying the wild-type pifA sequence, decreasing amounts of DNA
(corresponding to
62

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
10-fold dilutions from 104 to 10-7), were also included in the reaction to
estimate the
sensitivity. In these reactions only the primers ADV452 and ADV453 were used.
In a PCR
reaction with 30 cycles (10 cycles with annealing temperature of 53 C and an
additional 20
cycles with annealing temperature of 50 C), the sensitivity of the method was
1 in 100,000
(data not shown). As shown in figure 5, increasing the number of PCR cycles to
37 improved
the visual sensitivity of the method to 10-6 for the detection of D133V
revertants, without
significantly compromising the specificity. A clear band was visible at the 10-
6 dilution,
corresponding to a detection level of 1 copy of the wild-type sequence in a
million of the
D133V mutant, when 1 ng of plasmid was used as the initial amount of DNA. Only
a very
weak band can be visualized in lanes 1 and 9 after longer exposure, reassuring
the robust
specificity of the method. On the other hand, when starting with 5 ng of DNA,
a band could
be easily detected at the 10-7 dilution, increasing the sensitivity of the
PCR. However, a
similar band in intensity could also be detected with the pGG55 D133V plasmid,
indicating
the specificity limit of the method (Figure 17). This band observed with the
pGG55 D133V
plasmid is likely due to non-specific amplification of the D133V mutation with
primer
ADV452 that can significantly accumulate with the increased number of cycles.
These results
indicate that the sensitivity limit for this method, without significantly
compromising the
specificity, is situated between 1 to 1,000,000 and 1 to 10,000,000.
EXAMPLE 10: Recombinant Listeria expressing a fusion protein of LLO to E7(Lm-
LLO-E7)
11002461This strain is approx. 4 -5 logs more attenuated than the wild-type
parent strain
10403S and secretes the fusion protein tLLO-E7. This immunotherapy is based on
the
backbone XFL7, which is derived from 10403S by the irreversible deletion in
the virulence
gene transcription activator plfA. PrfA regulates the transcription of several
virulence genes
such as Listeriolysin 0 (LLO), ActA, PlcA (phospholipase A), PlcB
(phospholipase B) etc
that are required for in vivo intracellular growth and survival of L.
monocytogenes. The
plasmid pGG55 is retained by the Lm-LLO-E7 in vitro by means of selection with

`chloramphenica. However for in vivo retention of the plasmid by Lm-LLO-E7, it
carries a
copy of mutated prfA (D133V), which has been demonstrated to be less active
than wild-type
prfA in DNA binding and activating the transcription of virulence genes. We
have observed
that complementation with mutated prfA resulted in approx. 40 fold reduction
in the amount
of secreted LLO from Lm-LLO-E7 when compared to wild-type strain 10403S. This
implicates that possibly the strain Lm-LLO-E7 exhibits a reduced expression of
the virulence
63

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
genes that are regulated by prfA such as actA, in1A, in1B, in1C, plcB etc. In
Lm-LLO-E7, the
complementation with mutated copy of prfA possibly causes a reduction in the
expression of
different virulence genes that are regulated by prfA resulting in overall
attenuation of approx.
4-5 logs.
EXAMPLE 11: Immunotherapy with chemoradiation for anal cancer
11002471 Phase I-II Study
11002481 Eligibility Criteria
= Newly diagnosed locally advanced anal cancer:
= Stages: T > 4cm or node +.
= PS 0-1
= Staging by CT/MRI or PET scan.
= No significant cardiac or pulmonary disease. Adequate bone marrow and
renal function.
11002491 ADXS-HPV Treatment
= ADXS-HPV administered at lx109cfu
o IV Infusion of 500 ml over 30 minutes
= Premedications for ADXS-HPV treatment:
o Naprosyn 500 mg BID, Day -1, 0
o Promethazine 25 mg PO, BID (pre-dose, 8 hours)
= Post infusion antibiotics
o Ampicillin 500 mg QID, Days 3-9
[00250] Treatment schedule (see Figure 23-24).
11002511 Patients with newly diagnosed anal cancer with a primary tumor > 4cm
or lymph node
involvement, without distant metastases, are eligible. All patients receive 2
courses of
mitomycin, 5-FU with concurrent radiation (54 Gy in 30 fractions by intensity
modulated
radiation therapy [IMRT1). Patients receive 4 treatments of ADXS-HPV, 1x109
colony
forming units intravenously once approximately every 28 days. In treatment
schedule #1 (Fig.
23), the first dose is given before chemoradiation and the 2-4th doses are
given every 28 days
after completion of radiation. In treatment schedule #2 (Fig. 24), the second
dose of ADXS-
HPV is administered during chemoradiation.
RESULTS
64

CA 02964764 2017-04-13
WO 2016/061182
PCT/US2015/055462
[00252[8 patients have been treated on the locally advanced anal cancer trial.
Seven have had
completed treatment and have no evidence of disease (NED) (Table 1). (Only the
patient with
the grade 5 unrelated cardiac event is not alive and with NED. ¨ see Table 1)
[00253]
\\\sµ s\\ .\\ Z = kss,\N\
1 59 IIIB (T3N3) Yes CR, NED
Yes CR,
3 70 IIIA (T4NO) Yes CR, NED
-itmmmESS=:t.it.W(173N3YERYOMMEMENnUggMENnOgnMCfV19E1)MEMOR
Yes
4:ummun-71.-1 (4.-Sem,N01,Nontx--.r.etatedGt&CarclitovascularnPeridtrkgmumun
Munumu-:70=11tT3NE;MMN-Y-Petdgmmumu
mNmnmmmmmmmmmmmmmmmmmpmw:.n,::::mmmmm
[00254] Toxicities
= Rigors lasting for about 24 hours, often requiring meperidine, are most
common toxicity.
= No evidence of bacterial infection.
= No overlapping toxic ities with chemoradiation (1 grade 3 neutropenia).
[00255] While certain features of the invention have been illustrated and
described
herein, many modifications, substitutions, changes, and equivalents will now
occur to
those of ordinary skill in the art. It is, therefore, to be understood that
the appended claims
are intended to cover all such modifications and changes as fall within the
true spirit of
the invention.

Representative Drawing

Sorry, the representative drawing for patent document number 2964764 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2015-10-14
(87) PCT Publication Date 2016-04-21
(85) National Entry 2017-04-13
Dead Application 2022-01-04

Abandonment History

Abandonment Date Reason Reinstatement Date
2021-01-04 FAILURE TO REQUEST EXAMINATION
2021-04-14 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2017-04-13
Maintenance Fee - Application - New Act 2 2017-10-16 $100.00 2017-04-13
Maintenance Fee - Application - New Act 3 2018-10-15 $100.00 2018-09-17
Maintenance Fee - Application - New Act 4 2019-10-15 $100.00 2019-09-24
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Cover Page 2017-05-24 1 37
Abstract 2017-04-13 1 59
Claims 2017-04-13 17 562
Drawings 2017-04-13 22 1,289
Description 2017-04-13 65 3,775
Patent Cooperation Treaty (PCT) 2017-04-13 1 55
International Search Report 2017-04-13 11 721
National Entry Request 2017-04-13 5 200
Prosecution/Amendment 2017-04-13 6 197

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :